1
|
Moulton C, Baroni A, Quagliarini E, Leone L, Digiacomo L, Morotti M, Caracciolo G, Podda MV, Tasciotti E. Navigating the nano-bio immune interface: advancements and challenges in CNS nanotherapeutics. Front Immunol 2024; 15:1447567. [PMID: 39600701 PMCID: PMC11588692 DOI: 10.3389/fimmu.2024.1447567] [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: 06/12/2024] [Accepted: 10/21/2024] [Indexed: 11/29/2024] Open
Abstract
In recent years, significant advancements have been made in utilizing nanoparticles (NPs) to modulate immune responses within the central nervous system (CNS), offering new opportunities for nanotherapeutic interventions in neurological disorders. NPs can serve as carriers for immunomodulatory agents or platforms for delivering nucleic acid-based therapeutics to regulate gene expression and modulate immune responses. Several studies have demonstrated the efficacy of NP-mediated immune modulation in preclinical models of neurological diseases, including multiple sclerosis, stroke, Alzheimer's disease, and Parkinson's disease. While challenges remain, advancements in NPs engineering and design have led to the development of NPs using diverse strategies to overcome these challenges. The nano-bio interface with the immune system is key in the conceptualization of NPs to efficiently act as nanotherapeutics in the CNS. The biomolecular corona plays a pivotal role in dictating NPs behavior and immune recognition within the CNS, giving researchers the opportunity to optimize NPs design and surface modifications to minimize immunogenicity and enhance biocompatibility. Here, we review how NPs interact with the CNS immune system, focusing on immunosurveillance of NPs, NP-induced immune reprogramming and the impact of the biomolecular corona on NPs behavior in CNS immune responses. The integration of NPs into CNS nanotherapeutics offers promising opportunities for addressing the complex challenges of acute and chronic neurological conditions and pathologies, also in the context of preventive and rehabilitative medicine. By harnessing the nano-bio immune interface and understanding the significance of the biomolecular corona, researchers can develop targeted, safe, and effective nanotherapeutic interventions for a wide range of CNS disorders to improve treatment and rehabilitation. These advancements have the potential to revolutionize the treatment landscape of neurological diseases, offering promising solutions for improved patient care and quality of life in the future.
Collapse
Affiliation(s)
| | - Anna Baroni
- Human Longevity Program, IRCCS San Raffaele Roma, Rome, Italy
| | - Erica Quagliarini
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Lucia Leone
- Department of Neuroscience, Università Cattolica del Sacro Cuore, Rome, Italy
- Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Luca Digiacomo
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Marta Morotti
- Department of Neuroscience, Università Cattolica del Sacro Cuore, Rome, Italy
- Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Giulio Caracciolo
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Maria Vittoria Podda
- Department of Neuroscience, Università Cattolica del Sacro Cuore, Rome, Italy
- Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Ennio Tasciotti
- Human Longevity Program, IRCCS San Raffaele Roma, Rome, Italy
- Department of Human Sciences and Quality of Life Promotion, Università telematica San Raffaele, Rome, Italy
| |
Collapse
|
2
|
Zhou G, Li R, Sheng S, Huang J, Zhou F, Wei Y, Liu H, Su J. Organoids and organoid extracellular vesicles-based disease treatment strategies. J Nanobiotechnology 2024; 22:679. [PMID: 39506799 PMCID: PMC11542470 DOI: 10.1186/s12951-024-02917-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Accepted: 10/09/2024] [Indexed: 11/08/2024] Open
Abstract
Organoids are "mini-organs" that self-organize and differentiate from stem cells under in vitro 3D culture conditions, mimicking the spatial structure and function of tissues in vivo. Extracellular vesicles (EVs) are nanoscale phospholipid bilayer vesicles secreted by living cells, rich in bioactive molecules, with excellent biocompatibility and low immunogenicity. Compared to EVs, organoid-derived EVs (OEVs) exhibit higher yield and enhanced biological functions. Organoids possess stem cell characteristics, and OEVs are capable of delivering active substances, making both highly promising for medical applications. In this review, we provide an overview of the fundamental biological principles of organoids and OEVs, and discuss their current applications in disease treatment. We then focus on the differences between OEVs and traditional EVs. Subsequently, we present methods for the engineering modification of OEVs. Finally, we critically summarize the advantages and challenges of organoids and OEVs. In conclusion, we believe that a deeper understanding of organoids and OEVs will provide innovative solutions to complex diseases.
Collapse
Affiliation(s)
- Guangyin Zhou
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- Organoid Research Center, Shanghai University, Shanghai, 200444, China
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai, 200444, China
| | - Ruiyang Li
- Organoid Research Center, Shanghai University, Shanghai, 200444, China
- Department of Orthopedics, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Shihao Sheng
- Department of Orthopedics, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Jingtao Huang
- Department of Orthopedics, Shanghai Zhongye Hospital, Shanghai, 200444, China
| | - Fengjin Zhou
- Department of Orthopaedics, Honghui Hospital, Xi'an Jiao Tong University, Xi'an, 710000, China.
| | - Yan Wei
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China.
- Organoid Research Center, Shanghai University, Shanghai, 200444, China.
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai, 200444, China.
| | - Han Liu
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China.
- Organoid Research Center, Shanghai University, Shanghai, 200444, China.
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai, 200444, China.
- Department of Orthopedics, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China.
| | - Jiacan Su
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China.
- Organoid Research Center, Shanghai University, Shanghai, 200444, China.
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai, 200444, China.
- Department of Orthopedics, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China.
| |
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
|
Qi Z, Peng J, Wang H, Wang L, Su Y, Ding L, Cao B, Zhao Y, Xing Q, Yang J. Modulating neuroinflammation and cognitive function in postoperative cognitive dysfunction via CCR5-GPCRs-Ras-MAPK pathway targeting with microglial EVs. CNS Neurosci Ther 2024; 30:e14924. [PMID: 39143678 PMCID: PMC11324532 DOI: 10.1111/cns.14924] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2024] [Revised: 07/28/2024] [Accepted: 08/02/2024] [Indexed: 08/16/2024] Open
Abstract
AIMS Postoperative cognitive dysfunction (POCD) is prevalent among the elderly, characterized primarily by cognitive decline after surgery. This study aims to explore how extracellular vesicles (EVs) derived from BV2 microglial cells, with and without the C-C chemokine receptor type 5 (CCR5), affect neuroinflammation, neuronal integrity, and cognitive function in a POCD mouse model. METHODS We collected EVs from LPS-stimulated BV2 cells expressing CCR5 (EVsM1) and from BV2 cells with CCR5 knockdown (EVsM1-CCR5). These were administered to POCD-induced mice. Protein interactions between CCR5, G-protein-coupled receptors (GPCRs), and Ras were analyzed using structure-based docking and co-immunoprecipitation (Co-IP). We assessed the phosphorylation of p38 and Erk, the expression of synaptic proteins PSD95 and MAP2, and conducted Morris Water Maze tests to evaluate cognitive function. RESULTS Structure-based docking and Co-IP confirmed interactions between CCR5, GPR, and Ras, suggesting a CCR5-GPCRs-Ras-MAPK pathway involvement in neuroinflammation. EVsM1 heightened neuroinflammation, reduced synaptic integrity, and impaired cognitive function in POCD mice. In contrast, EVsM1-CCR5 reduced neuroinflammatory markers, preserved synaptic proteins, enhanced dendritic spine structure, and improved cognitive outcomes. CONCLUSION EVsM1 induced neuroinflammation via the CCR5-GPCRs-Ras-MAPK pathway, with EVsM1-CCR5 showing protective effects on POCD progression, suggesting a new therapeutic strategy for POCD management via targeted modification of microglial EVs.
Collapse
Affiliation(s)
- Zheng Qi
- Department of Anesthesiology, Pain and Perioperative MedicineThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Junlin Peng
- Department of Anesthesiology, Pain and Perioperative MedicineThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Haitao Wang
- Department of Anesthesiology, Pain and Perioperative MedicineThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Li Wang
- Biobank of The First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Yu Su
- Institutes of Biomedical SciencesFudan UniversityShanghaiChina
| | - Lan Ding
- Department of NeurologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Bin Cao
- Department of Anesthesiology, Pain and Perioperative MedicineThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Yingying Zhao
- Department of Anesthesiology, Pain and Perioperative MedicineThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Qinghe Xing
- Institutes of Biomedical SciencesFudan UniversityShanghaiChina
| | - Jian‐jun Yang
- Department of Anesthesiology, Pain and Perioperative MedicineThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| |
Collapse
|
5
|
Baudo G, Flinn H, Holcomb M, Tiwari A, Soriano S, Taraballi F, Godin B, Zinger A, Villapol S. Sex-dependent improvement in traumatic brain injury outcomes after liposomal delivery of dexamethasone in mice. Bioeng Transl Med 2024; 9:e10647. [PMID: 39036088 PMCID: PMC11256133 DOI: 10.1002/btm2.10647] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2023] [Revised: 12/02/2023] [Accepted: 01/11/2024] [Indexed: 07/23/2024] Open
Abstract
Traumatic brain injury (TBI) can have long-lasting physical, emotional, and cognitive consequences due to the neurodegeneration caused by its robust inflammatory response. Despite advances in rehabilitation care, effective neuroprotective treatments for TBI patients are lacking. Furthermore, current drug delivery methods for TBI treatment are inefficient in targeting inflamed brain areas. To address this issue, we have developed a liposomal nanocarrier (Lipo) encapsulating dexamethasone (Dex), an agonist for the glucocorticoid receptor utilized to alleviate inflammation and swelling in various conditions. In vitro studies show that Lipo-Dex were well tolerated in human and murine neural cells. Lipo-Dex showed significant suppression of inflammatory cytokines, IL-6 and TNF-α, release after induction of neural inflammation with lipopolysaccharide. Further, the Lipo-Dex were administered to young adult male and female C57BL/6 mice immediately after controlled cortical impact injury (a TBI model). Our findings demonstrate that Lipo-Dex can selectively target the injured brain, thereby reducing lesion volume, cell death, astrogliosis, the release of pro-inflammatory cytokines, and microglial activation compared to Lipo-treated mice in a sex-dependent manner, showing a major impact only in male mice. This highlights the importance of considering sex as a crucial variable in developing and evaluating new nano-therapies for brain injury. These results suggest that Lipo-Dex administration may effectively treat acute TBI.
Collapse
Affiliation(s)
- Gherardo Baudo
- Department of NanomedicineHouston Methodist Research InstituteHoustonTexasUSA
| | - Hannah Flinn
- Department of Neurosurgery and Center for NeuroregenerationHouston Methodist Research InstituteHoustonTexasUSA
| | - Morgan Holcomb
- Department of Neurosurgery and Center for NeuroregenerationHouston Methodist Research InstituteHoustonTexasUSA
| | - Anjana Tiwari
- Department of NanomedicineHouston Methodist Research InstituteHoustonTexasUSA
| | - Sirena Soriano
- Department of Neurosurgery and Center for NeuroregenerationHouston Methodist Research InstituteHoustonTexasUSA
| | - Francesca Taraballi
- Department of Orthopedics and Sports Medicine and Center for Musculoskeletal RegenerationHouston Methodist HospitalHoustonTexasUSA
| | - Biana Godin
- Department of NanomedicineHouston Methodist Research InstituteHoustonTexasUSA
- Department of Obstetrics and GynecologyHouston Methodist Research InstituteHoustonTexasUSA
- Department of Obstetrics and GynecologyWeill Cornell Medicine College (WCMC)New YorkNew YorkUSA
- Department of Biomedical EngineeringTexas A&M UniversityCollege StationTexasUSA
| | - Assaf Zinger
- Department of Cardiovascular SciencesHouston Methodist Research InstituteHoustonTexasUSA
- Department of Chemical EngineeringTechnion−Israel Institute of TechnologyHaifaIsrael
| | - Sonia Villapol
- Department of Neurosurgery and Center for NeuroregenerationHouston Methodist Research InstituteHoustonTexasUSA
- Department of Neuroscience in Neurological SurgeryWeill Cornell Medicine College (WCMC)New YorkNew YorkUSA
| |
Collapse
|
6
|
Liu H, Su J. Organoid extracellular vesicle-based therapeutic strategies for bone therapy. BIOMATERIALS TRANSLATIONAL 2023; 4:199-212. [PMID: 38282702 PMCID: PMC10817793 DOI: 10.12336/biomatertransl.2023.04.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/30/2023] [Revised: 11/24/2023] [Accepted: 12/05/2023] [Indexed: 01/30/2024]
Abstract
With the rapid development of population ageing, bone-related diseases seriously affecting the life of the elderly. Over the past few years, organoids, cell clusters with specific functions and structures that are self-induced from stem cells after three-dimensional culture in vitro, have been widely used for bone therapy. Moreover, organoid extracellular vesicles (OEVs) have emerging as promising cell-free nanocarriers due to their vigoroso physiological effects, significant biological functions, stable loading capacity, and great biocompatibility. In this review, we first provide a comprehensive overview of biogenesis, internalisation, isolation, and characterisation of OEVs. We then comprehensively highlight the differences between OEVs and traditional EVs. Subsequently, we present the applications of natural OEVs in disease treatment. We also summarise the engineering modifications of OEVs, including engineering parental cells and engineering OEVs after isolation. Moreover, we provide an outlook on the potential of natural and engineered OEVs in bone-related diseases. Finally, we critically discuss the advantages and challenges of OEVs in the treatment of bone diseases. We believe that a comprehensive discussion of OEVs will provide more innovative and efficient solutions for complex bone diseases.
Collapse
Affiliation(s)
- Han Liu
- Institute of Translational Medicine, Shanghai University, Shanghai, China
- Organoid Research Center, Shanghai University, Shanghai, China
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai, China
- Department of Orthopedics, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jiacan Su
- Institute of Translational Medicine, Shanghai University, Shanghai, China
- Organoid Research Center, Shanghai University, Shanghai, China
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai, China
- Department of Orthopedics, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
7
|
Sela M, Poley M, Mora-Raimundo P, Kagan S, Avital A, Kaduri M, Chen G, Adir O, Rozencweig A, Weiss Y, Sade O, Leichtmann-Bardoogo Y, Simchi L, Aga-Mizrachi S, Bell B, Yeretz-Peretz Y, Zaid Or A, Choudhary A, Rosh I, Cordeiro D, Cohen-Adiv S, Berdichevsky Y, Odeh A, Shklover J, Shainsky-Roitman J, Schroeder JE, Hershkovitz D, Hasson P, Ashkenazi A, Stern S, Laviv T, Ben-Zvi A, Avital A, Ashery U, Maoz BM, Schroeder A. Brain-Targeted Liposomes Loaded with Monoclonal Antibodies Reduce Alpha-Synuclein Aggregation and Improve Behavioral Symptoms in Parkinson's Disease. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023; 35:e2304654. [PMID: 37753928 PMCID: PMC7615408 DOI: 10.1002/adma.202304654] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 09/04/2023] [Indexed: 09/28/2023]
Abstract
Monoclonal antibodies (mAbs) hold promise in treating Parkinson's disease (PD), although poor delivery to the brain hinders their therapeutic application. In the current study, it is demonstrated that brain-targeted liposomes (BTL) enhance the delivery of mAbs across the blood-brain-barrier (BBB) and into neurons, thereby allowing the intracellular and extracellular treatment of the PD brain. BTL are decorated with transferrin to improve brain targeting through overexpressed transferrin-receptors on the BBB during PD. BTL are loaded with SynO4, a mAb that inhibits alpha-synuclein (AS) aggregation, a pathological hallmark of PD. It is shown that 100-nm BTL cross human BBB models intact and are taken up by primary neurons. Within neurons, SynO4 is released from the nanoparticles and bound to its target, thereby reducing AS aggregation, and enhancing neuronal viability. In vivo, intravenous BTL administration results in a sevenfold increase in mAbs in brain cells, decreasing AS aggregation and neuroinflammation. Treatment with BTL also improve behavioral motor function and learning ability in mice, with a favorable safety profile. Accordingly, targeted nanotechnologies offer a valuable platform for drug delivery to treat brain neurodegeneration.
Collapse
Affiliation(s)
- Mor Sela
- The Louis Family Laboratory for Targeted Drug Delivery and Personalized Medicine Technologies, Department of Chemical Engineering, Technion – Israel Institute of Technology, Haifa 32000, Israel
| | - Maria Poley
- The Louis Family Laboratory for Targeted Drug Delivery and Personalized Medicine Technologies, Department of Chemical Engineering, Technion – Israel Institute of Technology, Haifa 32000, Israel
| | - Patricia Mora-Raimundo
- The Louis Family Laboratory for Targeted Drug Delivery and Personalized Medicine Technologies, Department of Chemical Engineering, Technion – Israel Institute of Technology, Haifa 32000, Israel
| | - Shaked Kagan
- The Louis Family Laboratory for Targeted Drug Delivery and Personalized Medicine Technologies, Department of Chemical Engineering, Technion – Israel Institute of Technology, Haifa 32000, Israel
| | - Aviram Avital
- The Norman Seiden Multidisciplinary Program for Nanoscience and Nanotechnology, Technion – Israel Institute of Technology, Haifa 32000, Israel
| | - Maya Kaduri
- The Louis Family Laboratory for Targeted Drug Delivery and Personalized Medicine Technologies, Department of Chemical Engineering, Technion – Israel Institute of Technology, Haifa 32000, Israel
| | - Gal Chen
- The Louis Family Laboratory for Targeted Drug Delivery and Personalized Medicine Technologies, Department of Chemical Engineering, Technion – Israel Institute of Technology, Haifa 32000, Israel
- The Interdisciplinary Program for Biotechnology, Technion - Israel Institute of Technology, Haifa 32000, Israel
| | - Omer Adir
- The Norman Seiden Multidisciplinary Program for Nanoscience and Nanotechnology, Technion – Israel Institute of Technology, Haifa 32000, Israel
| | - Adi Rozencweig
- The Louis Family Laboratory for Targeted Drug Delivery and Personalized Medicine Technologies, Department of Chemical Engineering, Technion – Israel Institute of Technology, Haifa 32000, Israel
| | - Yfat Weiss
- School of Neurobiology, Biochemistry and Biophysics, George S. Wise Faculty of Life Sciences, Sagol School of Neuroscience, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Ofir Sade
- School of Neurobiology, Biochemistry and Biophysics, George S. Wise Faculty of Life Sciences, Sagol School of Neuroscience, Tel Aviv University, Tel Aviv 6997801, Israel
| | | | - Lilach Simchi
- Department of Occupational Therapy, Faculty of Social Welfare and Health Sciences, University of Haifa, Haifa 3498838, Israel
| | - Shlomit Aga-Mizrachi
- Department of Occupational Therapy, Faculty of Social Welfare and Health Sciences, University of Haifa, Haifa 3498838, Israel
| | - Batia Bell
- Department of Developmental Biology and Cancer Research, The Institute for Medical Research Israel-Canada, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem 9190500, Israel
| | - Yoel Yeretz-Peretz
- Department of Developmental Biology and Cancer Research, The Institute for Medical Research Israel-Canada, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem 9190500, Israel
| | - Aviv Zaid Or
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv 6997801, Israel
- Department of Physiology and Pharmacology, Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Ashwani Choudhary
- Sagol Department of Neurobiology, Faculty of Natural Sciences, University of Haifa, Haifa 3498838, Israel
| | - Idan Rosh
- Sagol Department of Neurobiology, Faculty of Natural Sciences, University of Haifa, Haifa 3498838, Israel
| | - Diogo Cordeiro
- Sagol Department of Neurobiology, Faculty of Natural Sciences, University of Haifa, Haifa 3498838, Israel
| | - Stav Cohen-Adiv
- The Department of Cell and Developmental Biology, Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Yevgeny Berdichevsky
- The Department of Cell and Developmental Biology, Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Anas Odeh
- Department of Genetics and Developmental Biology, The Rappaport Faculty of Medicine and Research Institute, Technion – Israel Institute of Technology, Haifa 32000, Israel
| | - Jeny Shklover
- The Louis Family Laboratory for Targeted Drug Delivery and Personalized Medicine Technologies, Department of Chemical Engineering, Technion – Israel Institute of Technology, Haifa 32000, Israel
| | - Janna Shainsky-Roitman
- The Louis Family Laboratory for Targeted Drug Delivery and Personalized Medicine Technologies, Department of Chemical Engineering, Technion – Israel Institute of Technology, Haifa 32000, Israel
| | - Joshua E. Schroeder
- Spine Unit, Orthopedic Complex, Hadassah Hebrew University Medical Center, Kiryat Hadassah, POB 12000, Jerusalem 9190500, Israel
| | - Dov Hershkovitz
- Department of Pathology, Tel Aviv Sourasky Medical Center, Tel Aviv 6997801, Israel
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Peleg Hasson
- Department of Genetics and Developmental Biology, The Rappaport Faculty of Medicine and Research Institute, Technion – Israel Institute of Technology, Haifa 32000, Israel
| | - Avraham Ashkenazi
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv 6997801, Israel
- The Department of Cell and Developmental Biology, Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Shani Stern
- Sagol Department of Neurobiology, Faculty of Natural Sciences, University of Haifa, Haifa 3498838, Israel
| | - Tal Laviv
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv 6997801, Israel
- Department of Physiology and Pharmacology, Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Ayal Ben-Zvi
- Department of Developmental Biology and Cancer Research, The Institute for Medical Research Israel-Canada, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem 9190500, Israel
| | - Avi Avital
- Department of Occupational Therapy, Faculty of Social Welfare and Health Sciences, University of Haifa, Haifa 3498838, Israel
| | - Uri Ashery
- School of Neurobiology, Biochemistry and Biophysics, George S. Wise Faculty of Life Sciences, Sagol School of Neuroscience, Tel Aviv University, Tel Aviv 6997801, Israel
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Ben M. Maoz
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv 6997801, Israel
- Department of Biomedical Engineering, Tel Aviv University, Tel Aviv 6997801, Israel
- The Center for Nanoscience and Nanotechnology, Tel Aviv University, Tel Aviv 6997801, Israel
- Sagol Center for Regenerative Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Avi Schroeder
- The Louis Family Laboratory for Targeted Drug Delivery and Personalized Medicine Technologies, Department of Chemical Engineering, Technion – Israel Institute of Technology, Haifa 32000, Israel
| |
Collapse
|
8
|
Qi Z, Yu Y, Su Y, Cao B, Shao H, Yang JJ. M1-Type Microglia-Derived Extracellular Vesicles Overexpressing IL-1R1 Promote Postoperative Cognitive Dysfunction by Regulating Neuronal Inflammation. Inflammation 2023; 46:2254-2269. [PMID: 37505422 DOI: 10.1007/s10753-023-01875-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 06/30/2023] [Accepted: 07/10/2023] [Indexed: 07/29/2023]
Abstract
Postoperative cognitive dysfunction (POCD) is a common complication after surgical anesthesia, mainly manifested as memory impairment, decreased attention, and cognitive function with mood and personality changes. Activated microglia (M1-type microglia) have been demonstrated to release inflammatory substances (IL-1β, TNF-α, etc.) that cause neuronal degeneration and death by activating the NF-κB signaling pathway and upregulating Caspase-3 and Bax. However, the pathogenesis of POCD is still not fully understood and needs further research. In the present study, we investigated the effect of M1-type microglia-derived extracellular vesicles (EVsM1-Microglia) in the pathological process of POCD. The levels of NF-κB phosphorylation and IL-1β protein expression in hippocampal neurons were significantly increased in the Surgery group, while PSD95 and MAP2 were significantly decreased. Surgery induced microglia activation, synapse-associated protein decrease, and neuronal degeneration in hippocampus. And the amount of spine and mushroom spine significantly decreased in surgical mice, which was reverted in the presence of IL-1R1 siRNA. In addition, EVsM1-Microglia promoted synaptic loss and neuron degeneration independent of surgery and microglia activation. Furthermore, EVsM1-Microglia promoted memory defects in surgical mice. We demonstrated that EVsM1-Microglia with high expression of IL-1R1 promote POCD development by regulating neuronal inflammation.
Collapse
Affiliation(s)
- Zheng Qi
- Department of Anesthesiology, Pain and Perioperative Medicine, The First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe Dong Road, Zhengzhou, 450000, People's Republic of China
- Henan Province International Joint Laboratory of Pain, Cognition and Emotion, The First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe Dong Road, Zhengzhou, 450000, People's Republic of China
| | - Yang Yu
- Department of Anesthesiology, Pain and Perioperative Medicine, The First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe Dong Road, Zhengzhou, 450000, People's Republic of China
- Henan Province International Joint Laboratory of Pain, Cognition and Emotion, The First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe Dong Road, Zhengzhou, 450000, People's Republic of China
| | - Yu Su
- Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, People's Republic of China
| | - Bin Cao
- Department of Anesthesiology, Pain and Perioperative Medicine, The First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe Dong Road, Zhengzhou, 450000, People's Republic of China
- Henan Province International Joint Laboratory of Pain, Cognition and Emotion, The First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe Dong Road, Zhengzhou, 450000, People's Republic of China
| | - Hua Shao
- Department of Anesthesiology, Pain and Perioperative Medicine, The First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe Dong Road, Zhengzhou, 450000, People's Republic of China
- Henan Province International Joint Laboratory of Pain, Cognition and Emotion, The First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe Dong Road, Zhengzhou, 450000, People's Republic of China
| | - Jian-Jun Yang
- Department of Anesthesiology, Pain and Perioperative Medicine, The First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe Dong Road, Zhengzhou, 450000, People's Republic of China.
- Henan Province International Joint Laboratory of Pain, Cognition and Emotion, The First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe Dong Road, Zhengzhou, 450000, People's Republic of China.
| |
Collapse
|
9
|
Zhang L, Liu J, Zhou C. Current aspects of small extracellular vesicles in pain process and relief. Biomater Res 2023; 27:78. [PMID: 37563666 PMCID: PMC10416402 DOI: 10.1186/s40824-023-00417-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Accepted: 08/04/2023] [Indexed: 08/12/2023] Open
Abstract
Small extracellular vesicles (sEVs) have been identified as a noteworthy paracrine mechanism of intercellular communication in diagnosing and managing neurological disorders. Current research suggests that sEVs play a pivotal role in the pathological progression of pain, emphasizing their critical function in the pathological progression of pain in acute and chronic pain models. By facilitating the transfer of diverse molecules, such as proteins, nucleic acids, and metabolites, sEVs can modulate pain signaling transmission in both the central and peripheral nervous systems. Furthermore, the unique molecules conveyed by sEVs in pain disorders indicate their potential as diagnostic biomarkers. The application of sEVs derived from mesenchymal stem cells (MSCs) in regenerative pain medicine has emerged as a promising strategy for pain management. Moreover, modified sEVs have garnered considerable attention in the investigation of pathological processes and therapeutic interventions. This review presents a comprehensive overview of the current knowledge regarding the involvement of sEVs in pain pathogenesis and treatment. Nevertheless, additional research is imperative to facilitate their clinical implementation. Schematic diagram of sEVs in the biogenesis, signal transmission, diagnosis, and treatment of pain disorders. Small extracellular vesicles (sEVs) are secreted by multiple cells, loading with various biomolecules, such as miRNAs, transmembrane proteins, and amino acids. They selectively target other cells and regulating pain signal transmission. The composition of sEVs can serve as valuable biomarkers for pain diagnosis. In particular, mesenchymal stem cell-derived sEVs have shown promise as regenerative medicine for managing multiple pain disorders. Furthermore, by modifying the structure or contents of sEVs, they could potentially be used as a potent analgesic method.
Collapse
Affiliation(s)
- Lanyu Zhang
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, China
- Laboratory of Anesthesia & Critical Care Medicine, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology, West China Hospital of Sichuan University, Chengdu, 610041, Sichuan, China
| | - Jin Liu
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, China
| | - Cheng Zhou
- Laboratory of Anesthesia & Critical Care Medicine, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology, West China Hospital of Sichuan University, Chengdu, 610041, Sichuan, China.
| |
Collapse
|
10
|
Mougenot MF, Pereira VS, Costa ALR, Lancellotti M, Porcionatto MA, da Silveira JC, de la Torre LG. Biomimetic Nanovesicles—Sources, Design, Production Methods, and Applications. Pharmaceutics 2022; 14:pharmaceutics14102008. [PMID: 36297442 PMCID: PMC9610935 DOI: 10.3390/pharmaceutics14102008] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 09/19/2022] [Accepted: 09/20/2022] [Indexed: 11/18/2022] Open
Abstract
Despite all the progress in the field of liposomes and nanoparticles for applications as drug and gene delivery systems, the specific targeting and immune system escape capabilities of these systems are still limited. Biomimetic nanovesicles emerged as a strategy to overcome these and other limitations associated with synthetic carriers, such as short circulation time, cytotoxicity, and difficulty in crossing biological barriers, since many of the desirable abilities of drug delivery systems are innate characteristics of biological vesicles. Thus, the question arises: would biomimetic nanovesicles be responsible for addressing these advances? It is currently known that biomimetic nanovesicles (BNV) can combine the intrinsic advantages of natural materials with the well-known production methods and controllability of synthetic systems. Besides, the development of the biotechnology and nanotechnology fields has provided a better understanding of the functionalities of biological vesicles and the means for the design and production of biomimetic nanovesicles (BNV). Based on this, this work will focus on tracking the main research on biomimetic nanovesicles (BNV) applied as drug and gene delivery systems, and for vaccines applications. In addition, it will describe the different sources of natural vesicles, the technical perspectives on obtaining them, and the possibility of their hybridization with synthetic liposomes.
Collapse
Affiliation(s)
- Marcel Franco Mougenot
- Department of Materials and Bioprocesses Engineering, School of Chemical Engineering, University of Campinas, Campinas 13083-970, Brazil
| | - Vanessa Sousa Pereira
- Department of Materials and Bioprocesses Engineering, School of Chemical Engineering, University of Campinas, Campinas 13083-970, Brazil
| | - Ana Letícia Rodrigues Costa
- Department of Materials and Bioprocesses Engineering, School of Chemical Engineering, University of Campinas, Campinas 13083-970, Brazil
- Institute of Exact and Technological Sciences, Campus Florestal, Federal University of Viçosa (UFV), Florestal 35690-000, Brazil
| | - Marcelo Lancellotti
- Faculty of Pharmaceutical Sciences, University of Campinas (UNICAMP), Campinas 13083-871, Brazil
| | | | - Juliano Coelho da Silveira
- Department of Veterinary Medicine, Faculty of Animal Sciences and Food Engineering, University of São Paulo, Pirassununga 13635-900, Brazil
| | - Lucimara Gaziola de la Torre
- Department of Materials and Bioprocesses Engineering, School of Chemical Engineering, University of Campinas, Campinas 13083-970, Brazil
- Correspondence: ; Tel.: +55-19-3521-0397
| |
Collapse
|
11
|
Wang C, Wu S. Research update on cell membrane camouflaged nanoparticles for cancer therapy. Front Bioeng Biotechnol 2022; 10:944518. [PMID: 35992357 PMCID: PMC9388754 DOI: 10.3389/fbioe.2022.944518] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Accepted: 07/06/2022] [Indexed: 11/13/2022] Open
Abstract
Cell membrane-camouflaged biomimetic functionalization of nanoparticles has emerged as a promising strategy for cancer theranostics. These cell membranes used for camouflaging are generally isolated from natural or engineered erythrocytes, neutrophils, macrophages, T lymphatic cells, stem cells, and cancer cells. The camouflaging strategy of coating nanoparticles with cell membranes allows for tumor homotypic targeting through self-recognition as source cells, immune evasion, and a prolonged blood circulation time, thereby improving the effective payload delivery and tumor therapy. More so, some engineered cell membranes with functionalized peptides, proteins and moieties on membrane surface can be transferred for therapy in the same time. In this review, we summarize the latest research on various types of cell membrane-camouflaged nanoparticles aimed at anti-cancer therapy, focusing on the biological advantages of different cell membranes, constitutions of nanoparticles, fabrication processes, key findings, potential therapies, and discuss the major challenges and future opportunities.
Collapse
|
12
|
Carvalho BG, Ceccato BT, Michelon M, Han SW, de la Torre LG. Advanced Microfluidic Technologies for Lipid Nano-Microsystems from Synthesis to Biological Application. Pharmaceutics 2022; 14:141. [PMID: 35057037 PMCID: PMC8781930 DOI: 10.3390/pharmaceutics14010141] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Revised: 12/23/2021] [Accepted: 12/30/2021] [Indexed: 12/17/2022] Open
Abstract
Microfluidics is an emerging technology that can be employed as a powerful tool for designing lipid nano-microsized structures for biological applications. Those lipid structures can be used as carrying vehicles for a wide range of drugs and genetic materials. Microfluidic technology also allows the design of sustainable processes with less financial demand, while it can be scaled up using parallelization to increase production. From this perspective, this article reviews the recent advances in the synthesis of lipid-based nanostructures through microfluidics (liposomes, lipoplexes, lipid nanoparticles, core-shell nanoparticles, and biomimetic nanovesicles). Besides that, this review describes the recent microfluidic approaches to produce lipid micro-sized structures as giant unilamellar vesicles. New strategies are also described for the controlled release of the lipid payloads using microgels and droplet-based microfluidics. To address the importance of microfluidics for lipid-nanoparticle screening, an overview of how microfluidic systems can be used to mimic the cellular environment is also presented. Future trends and perspectives in designing novel nano and micro scales are also discussed herein.
Collapse
Affiliation(s)
- Bruna G. Carvalho
- Department of Material and Bioprocess Engineering, School of Chemical Engineering, University of Campinas (UNICAMP), Campinas 13083-852, Brazil; (B.G.C.); (B.T.C.)
| | - Bruno T. Ceccato
- Department of Material and Bioprocess Engineering, School of Chemical Engineering, University of Campinas (UNICAMP), Campinas 13083-852, Brazil; (B.G.C.); (B.T.C.)
| | - Mariano Michelon
- School of Chemical and Food Engineering, Federal University of Rio Grande (FURG), Rio Grande 96203-900, Brazil;
| | - Sang W. Han
- Center for Cell Therapy and Molecular, Department of Biophysics, Federal University of São Paulo (UNIFESP), São Paulo 04044-010, Brazil;
| | - Lucimara G. de la Torre
- Department of Material and Bioprocess Engineering, School of Chemical Engineering, University of Campinas (UNICAMP), Campinas 13083-852, Brazil; (B.G.C.); (B.T.C.)
| |
Collapse
|