1
|
Mu Z, Qin J, Zhou X, Wang K. Synergistic effects of human umbilical cord mesenchymal stem cells/neural stem cells and epidural electrical stimulation on spinal cord injury rehabilitation. Sci Rep 2024; 14:26090. [PMID: 39478010 PMCID: PMC11526023 DOI: 10.1038/s41598-024-75754-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: 07/16/2024] [Accepted: 10/08/2024] [Indexed: 11/02/2024] Open
Abstract
Spinal cord injury (SCI) is a severe neurological condition marked by a complex pathology leading to irreversible functional loss, which current treatments fail to improve. Epidural electrical stimulation (EES) shows promise in alleviating pathological pain, regulating hemodynamic disturbances, and enhancing motor function by modulating residual interneurons in the lower spinal cord. Cell transplantation (CT), especially using human umbilical cord mesenchymal stem cells (hUCMSCs) and neural stem cells (NSCs), has significantly improved sensory and motor recovery in SCI. However, the limitations of single treatments have driven the exploration of a multifaceted strategy, combining various modalities to optimize recovery at different stages. To comprehensively investigate the effectiveness of in situ transplantation of hUCMSCs/NSCs combined with subacute epidural electrical stimulation in a murine spinal cord crush injury model, providing valuable references for future animal studies and clinical research. In this study, we first examined neural stem cell changes via mRNA sequencing in an in vitro Transwell co-culture model. We then explored cell interaction mechanisms using proliferation assays, differentiation assays, and neuron complexity analysis. For animal experiments, 40 C57BL/6 mice were assigned to four groups (Injury/EES/CT/Combination). Histological evaluations employed HE and immunofluorescence staining, while electrophysiological and behavioral tests assessed motor recovery. Quantitative data were reported as mean ± standard error, with statistical analyses performed using GraphPad Prism and SPSS. Initially, we found that NSCs in the in vitro co-culture model showed a unique expression profile of differentially expressed genes (DEGs) compared to controls. GO/KEGG analysis indicated these DEGs were mainly linked to cell differentiation and growth factor secretion pathways. Neuronal and astrocytic markers further confirmed enhanced NSC differentiation and neuronal maturation in the co-culture model. In vivo, live imaging and human nuclei immunofluorescence staining revealed that transplanted cells persisted for some time post-transplantation. Histological analysis showed that during acute inflammation, both the stem cell and combined therapy groups significantly inhibited microglial polarization. In the chronic phase, these groups reduced fibrotic scar formation and encouraged astrocytic bridging. Behavioral tests, including swimming and gait analysis, demonstrated that combined CT and EES therapy was more effective than either treatment alone. In summary, the combined therapy offers a promising approach for spinal cord injury treatment, providing superior outcomes over individual treatments. Our findings underscore the potential of a combined treatment approach utilizing stem cells transplantation and EES as an effective strategy for the comprehensive management of spinal cord crush injury in mice. This integrated approach holds promise for enhancing functional recovery and improving the quality of life for individuals with spinal cord injury (SCI).
Collapse
Affiliation(s)
- Zhiping Mu
- Department of Bone and Joint Surgery, The Second Affiliated Hospital of Medical College, Xi'an Jiaotong University, Xi'an, 710004, Shaanxi, People's Republic of China
| | - Jiaodi Qin
- First Clinical Institute, Chongqing Medical University, Chongqing, 400016, People's Republic of China
| | - Xiaohua Zhou
- Chongqing Shizhu Tujia Autonomous County Traditional Chinese Medicine Hospital, Chongqing, 409199, People's Republic of China
| | - Kunzheng Wang
- Department of Bone and Joint Surgery, The Second Affiliated Hospital of Medical College, Xi'an Jiaotong University, Xi'an, 710004, Shaanxi, People's Republic of China.
| |
Collapse
|
2
|
Sato M, Inada E, Kubota N, Ozawa M. Loss of Cell-Cell Contact Inhibits Cellular Differentiation of α-Catenin Knock Out P19 Embryonal Carcinoma Cells and Their Colonization into the Developing Mouse Embryos. BIOTECH 2024; 13:41. [PMID: 39449371 DOI: 10.3390/biotech13040041] [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: 08/05/2024] [Revised: 09/17/2024] [Accepted: 09/24/2024] [Indexed: 10/26/2024] Open
Abstract
Cadherin-catenin cell-cell adhesion complexes, composed of cadherin, β-catenin or plakoglobin, and α-catenin (α-cat) molecules, are crucial for maintaining cell-cell contact and are commonly referred to as "adherens junctions (AJs)." Inactivating this system leads to loss of cell-cell contact and developmental arrest in early embryos. However, it remains unclear whether the loss of cell-cell contact affects the differentiation of embryonic cells. In this study, we explored the use of a murine embryonal carcinoma cell line, P19, as an in vitro model for early embryogenesis. P19 cells easily form embryoid bodies (EBs) and are susceptible to cellular differentiation in response to retinoic acid (RA) and teratoma formation. Using CRISPR/Cas9 technology to disrupt the endogenous α-cat gene in P19 cells, we generated α-cat knockout (KO) cells that exhibited a loss of cell-cell contact. When cultivated on non-coated dishes, these α-cat KO cells formed EBs, but their structures were labile. In the RA-containing medium, the α-cat KO EBs failed to produce differentiated cells on their outer layer and continued to express SSEA-1, an antigen specific to pluripotent cells. Teratoma formation assays revealed an absence of overt differentiated cells in tumors derived from α-cat KO P19 cells. Aggregation assays revealed the inability of the KO cells to colonize into the zona pellucida-denuded 8-cell embryos. These findings suggest that the AJs are essential for promoting the early stages of cellular differentiation and for the colonization of early-developing embryos.
Collapse
Affiliation(s)
- Masahiro Sato
- Section of Gene Expression Regulation, Frontier Science Research Center, Kagoshima University, Kagoshima 890-8544, Japan
| | - Emi Inada
- Department of Pediatric Dentistry, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima 890-8544, Japan
| | - Naoko Kubota
- Department of Pediatric Dentistry, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima 890-8544, Japan
| | - Masayuki Ozawa
- Department of Biochemistry and Molecular Biology, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima 890-8544, Japan
| |
Collapse
|
3
|
Gonsalves N, Sun MK, Chopra P, Latchoumane CF, Bajwa S, Tang R, Patel B, Boons GJ, Karumbaiah L. Neuritogenic glycosaminoglycan hydrogels promote functional recovery after severe traumatic brain injury. J Neural Eng 2024; 21:036058. [PMID: 38806019 PMCID: PMC11209949 DOI: 10.1088/1741-2552/ad5108] [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: 11/20/2023] [Revised: 04/22/2024] [Accepted: 05/28/2024] [Indexed: 05/30/2024]
Abstract
Objective.Severe traumatic brain injury (sTBI) induced neuronal loss and brain atrophy contribute significantly to long-term disabilities. Brain extracellular matrix (ECM) associated chondroitin sulfate (CS) glycosaminoglycans promote neural stem cell (NSC) maintenance, and CS hydrogel implants have demonstrated the ability to enhance neuroprotection, in preclinical sTBI studies. However, the ability of neuritogenic chimeric peptide (CP) functionalized CS hydrogels in promoting functional recovery, after controlled cortical impact (CCI) and suction ablation (SA) induced sTBI, has not been previously demonstrated. We hypothesized that neuritogenic (CS)CP hydrogels will promote neuritogenesis of human NSCs, and accelerate brain tissue repair and functional recovery in sTBI rats.Approach.We synthesized chondroitin 4-Osulfate (CS-A)CP, and 4,6-O-sulfate (CS-E)CP hydrogels, using strain promoted azide-alkyne cycloaddition (SPAAC), to promote cell adhesion and neuritogenesis of human NSCs,in vitro; and assessed the ability of (CS-A)CP hydrogels in promoting tissue and functional repair, in a novel CCI-SA sTBI model,in vivo. Main results.Results indicated that (CS-E)CP hydrogels significantly enhanced human NSC aggregation and migration via focal adhesion kinase complexes, when compared to NSCs in (CS-A)CP hydrogels,in vitro. In contrast, NSCs encapsulated in (CS-A)CP hydrogels differentiated into neurons bearing longer neurites and showed greater spontaneous activity, when compared to those in (CS-E)CP hydrogels. The intracavitary implantation of (CS-A)CP hydrogels, acutely after CCI-SA-sTBI, prevented neuronal and axonal loss, as determined by immunohistochemical analyses. (CS-A)CP hydrogel implanted animals also demonstrated the significantly accelerated recovery of 'reach-to-grasp' function when compared to sTBI controls, over a period of 5-weeks.Significance.These findings demonstrate the neuritogenic and neuroprotective attributes of (CS)CP 'click' hydrogels, and open new avenues for the development of multifunctional glycomaterials that are functionalized with biorthogonal handles for sTBI repair.
Collapse
Affiliation(s)
- Nathan Gonsalves
- Regenerative Bioscience Center, University of Georgia, Athens, GA, United States of America
- Division of Neuroscience, Biomedical and Translational Sciences Institute, University of Georgia, Athens, GA, United States of America
| | - Min Kyoung Sun
- Regenerative Bioscience Center, University of Georgia, Athens, GA, United States of America
- Division of Neuroscience, Biomedical and Translational Sciences Institute, University of Georgia, Athens, GA, United States of America
| | - Pradeep Chopra
- Complex Carbohydrate Research Center, University of Georgia, Athens, GA, United States of America
| | - Charles-Francois Latchoumane
- Regenerative Bioscience Center, University of Georgia, Athens, GA, United States of America
- Edgar L. Rhodes Center for Animal and Dairy Science, College of Agriculture and Environmental Sciences, University of Georgia, Athens, GA, United States of America
| | - Simar Bajwa
- Regenerative Bioscience Center, University of Georgia, Athens, GA, United States of America
| | - Ruiping Tang
- Regenerative Bioscience Center, University of Georgia, Athens, GA, United States of America
- Edgar L. Rhodes Center for Animal and Dairy Science, College of Agriculture and Environmental Sciences, University of Georgia, Athens, GA, United States of America
| | - Bianca Patel
- Regenerative Bioscience Center, University of Georgia, Athens, GA, United States of America
| | - Geert-Jan Boons
- Complex Carbohydrate Research Center, University of Georgia, Athens, GA, United States of America
- Department of Chemistry, University of Georgia, Athens, GA, United States of America
- Department of Chemical Biology and Drug Discovery, Utrecht Institute for Pharmaceutical Sciences, and Bijvoet Center for Biomolecular Research, Utrecht University, Utrecht, The Netherlands
| | - Lohitash Karumbaiah
- Regenerative Bioscience Center, University of Georgia, Athens, GA, United States of America
- Division of Neuroscience, Biomedical and Translational Sciences Institute, University of Georgia, Athens, GA, United States of America
- Edgar L. Rhodes Center for Animal and Dairy Science, College of Agriculture and Environmental Sciences, University of Georgia, Athens, GA, United States of America
| |
Collapse
|
4
|
Hudakova N, Mudronova D, Marcincakova D, Slovinska L, Majerova P, Maloveska M, Petrouskova P, Humenik F, Cizkova D. The role of primed and non-primed MSC-derived conditioned media in neuroregeneration. Front Mol Neurosci 2023; 16:1241432. [PMID: 38025267 PMCID: PMC10656692 DOI: 10.3389/fnmol.2023.1241432] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Accepted: 10/16/2023] [Indexed: 12/01/2023] Open
Abstract
Introduction With growing significance in nervous system repair, mesenchymal stem cell-derived conditioned media (MSCCM) have been used in cell-free therapies in regenerative medicine. However, the immunomodulatory and neuroregenerative effects of MSCCM and the influence of priming on these effects are still poorly understood. Methods In this study, by various methods focused on cell viability, proliferation, neuron-like differentiation, neurite outgrowth, cell migration and regrowth, we demonstrated that MSCCM derived from adipose tissue (AT-MSCCM) and amniotic membrane (AM-MSCCM) had different effects on SH-SY5Y cells. Results and discussion AT-MSCCM was found to have a higher proliferative capacity and the ability to impact neurite outgrowth during differentiation, while AM-MSCCM showed more pronounced immunomodulatory activity, migration, and re-growth of SH-SY5Y cells in the scratch model. Furthermore, priming of MSC with pro-inflammatory cytokine (IFN-γ) resulted in different proteomic profiles of conditioned media from both sources, which had the highest effect on SH-SY5Y proliferation and neurite outgrowth in terms of the length of neurites (pAT-MSCCM) compared to the control group (DMEM). Altogether, our results highlight the potential of primed and non-primed MSCCM as a therapeutic tool for neurodegenerative diseases, although some differences must be considered.
Collapse
Affiliation(s)
- Nikola Hudakova
- Centre of Experimental and Clinical Regenerative Medicine, University of Veterinary Medicine and Pharmacy in Kosice, Košice, Slovakia
| | - Dagmar Mudronova
- Department of Microbiology and Immunology, University of Veterinary Medicine and Pharmacy in Kosice, Košice, Slovakia
| | - Dana Marcincakova
- Department of Pharmacology and Toxicology, University of Veterinary Medicine and Pharmacy in Kosice, Košice, Slovakia
| | - Lucia Slovinska
- Associated Tissue Bank, Faculty of Medicine, Pavol Jozef Safarik University and Luis Pasteur University Hospital, Košice, Slovakia
| | - Petra Majerova
- Institute of Neuroimmunology, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Marcela Maloveska
- Centre of Experimental and Clinical Regenerative Medicine, University of Veterinary Medicine and Pharmacy in Kosice, Košice, Slovakia
| | - Patricia Petrouskova
- Centre of Experimental and Clinical Regenerative Medicine, University of Veterinary Medicine and Pharmacy in Kosice, Košice, Slovakia
| | - Filip Humenik
- Centre of Experimental and Clinical Regenerative Medicine, University of Veterinary Medicine and Pharmacy in Kosice, Košice, Slovakia
| | - Dasa Cizkova
- Centre of Experimental and Clinical Regenerative Medicine, University of Veterinary Medicine and Pharmacy in Kosice, Košice, Slovakia
- Institute of Neuroimmunology, Slovak Academy of Sciences, Bratislava, Slovakia
| |
Collapse
|
5
|
Baglamis S, Sheraton VM, Meijer D, Qian H, Hoebe RA, Lenos KJ, Betjes MA, Betjes MA, Tans S, van Zon J, Vermeulen L, Krawczyk PM. Using picoliter droplet deposition to track clonal competition in adherent and organoid cancer cell cultures. Sci Rep 2023; 13:18832. [PMID: 37914743 PMCID: PMC10620187 DOI: 10.1038/s41598-023-42849-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: 02/12/2023] [Accepted: 09/15/2023] [Indexed: 11/03/2023] Open
Abstract
Clonal growth and competition underlie processes of key relevance in etiology, progression and therapy response across all cancers. Here, we demonstrate a novel experimental approach, based on multi-color, fluorescent tagging of cell nuclei, in combination with picoliter droplet deposition, to study the clonal dynamics in two- and three-dimensional cell cultures. The method allows for the simultaneous visualization and analysis of multiple clones in individual multi-clonal colonies, providing a powerful tool for studying clonal dynamics and identifying clonal populations with distinct characteristics. Results of our experiments validate the utility of the method in studying clonal dynamics in vitro, and reveal differences in key aspects of clonal behavior of different cancer cell lines in monoculture conditions, as well as in co-cultures with stromal fibroblasts.
Collapse
Affiliation(s)
- Selami Baglamis
- Laboratory for Experimental Oncology and Radiobiology, Center for Experimental and Molecular Medicine, Amsterdam UMC Location University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
- Cancer Center Amsterdam, 1081 HV, Amsterdam, The Netherlands
- Oncode Institute, 3521 AL, Utrecht, The Netherlands
- Amsterdam Gastroenterology Endocrinology Metabolism, 1105 AZ, Amsterdam, The Netherlands
| | - Vivek M Sheraton
- Laboratory for Experimental Oncology and Radiobiology, Center for Experimental and Molecular Medicine, Amsterdam UMC Location University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
- Cancer Center Amsterdam, 1081 HV, Amsterdam, The Netherlands
- Oncode Institute, 3521 AL, Utrecht, The Netherlands
- Amsterdam Gastroenterology Endocrinology Metabolism, 1105 AZ, Amsterdam, The Netherlands
- Institute for Advanced Study, University of Amsterdam, 1012 WX, Amsterdam, The Netherlands
| | - Debora Meijer
- Cancer Center Amsterdam, 1081 HV, Amsterdam, The Netherlands
- Department of Medical Biology, Amsterdam University Medical Centers (location AMC), University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
| | - Haibin Qian
- Cancer Center Amsterdam, 1081 HV, Amsterdam, The Netherlands
- Department of Medical Biology, Amsterdam University Medical Centers (location AMC), University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
| | - Ron A Hoebe
- Cancer Center Amsterdam, 1081 HV, Amsterdam, The Netherlands
- Department of Medical Biology, Amsterdam University Medical Centers (location AMC), University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
| | - Kristiaan J Lenos
- Laboratory for Experimental Oncology and Radiobiology, Center for Experimental and Molecular Medicine, Amsterdam UMC Location University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
- Cancer Center Amsterdam, 1081 HV, Amsterdam, The Netherlands
- Oncode Institute, 3521 AL, Utrecht, The Netherlands
- Amsterdam Gastroenterology Endocrinology Metabolism, 1105 AZ, Amsterdam, The Netherlands
| | - Max A Betjes
- Laboratory for Experimental Oncology and Radiobiology, Center for Experimental and Molecular Medicine, Amsterdam UMC Location University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
- Cancer Center Amsterdam, 1081 HV, Amsterdam, The Netherlands
- Oncode Institute, 3521 AL, Utrecht, The Netherlands
- Amsterdam Gastroenterology Endocrinology Metabolism, 1105 AZ, Amsterdam, The Netherlands
| | | | | | | | - Louis Vermeulen
- Laboratory for Experimental Oncology and Radiobiology, Center for Experimental and Molecular Medicine, Amsterdam UMC Location University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands.
- Cancer Center Amsterdam, 1081 HV, Amsterdam, The Netherlands.
- Oncode Institute, 3521 AL, Utrecht, The Netherlands.
- Amsterdam Gastroenterology Endocrinology Metabolism, 1105 AZ, Amsterdam, The Netherlands.
| | - Przemek M Krawczyk
- Cancer Center Amsterdam, 1081 HV, Amsterdam, The Netherlands.
- Department of Medical Biology, Amsterdam University Medical Centers (location AMC), University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands.
| |
Collapse
|
6
|
Daou B, Silvestri A, Lasa H, Mancino D, Prato M, Alegret N. Organic Functional Group on Carbon Nanotube Modulates the Maturation of SH-SY5Y Neuronal Models. Macromol Biosci 2023; 23:e2300173. [PMID: 37392465 DOI: 10.1002/mabi.202300173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Revised: 06/29/2023] [Accepted: 06/29/2023] [Indexed: 07/03/2023]
Abstract
Carbon nanotubes (CNT) have proven to be excellent substrates for neuronal cultures, showing high affinity and greatly boosting their synaptic functionality. Therefore, growing cells on CNT offers an opportunity to perform a large variety of neuropathology studies in vitro. To date, the interactions between neurons and chemical functional groups have not been studied extensively. To this end, multiwalled CNT (f-CNT) is functionalized with various functional groups, including sulfonic (-SO3 H), nitro (-NO2 ), amino (-NH2 ), and oxidized moieties. f-CNTs are spray-coated onto untreated glass substrates and are used as substrates for the incubation of neuroblastoma cells (SH-SY5Y). After 7 d, its effect is evaluated in terms of cell attachment, survival, growth, and spontaneous differentiation. Cell viability assays show quite increased proliferation on various f-CNT substrates (CNTs-NO2 > ox-CNTs ≈ CNTs-SO3 H > CNTs ≈ CNTs-NH2 ). Additionally, SH-SY5Y cells show selectively better differentiation and maturation with -SO3 H substrates, where an increased expression of β-III tubulin is seen. In all cases, intricate cell-CNT networks are observed and the morphology of the cells adopts longer and thinner cellular processes, suggesting that the type of functionalization may have an effect of the length and thickness. Finally, a possible correlation is determined between conductivity of f-CNTs and cell-processes lengths.
Collapse
Affiliation(s)
- Bahaa Daou
- Center for Cooperative Research in Biomaterials (CIC BiomaGUNE), Basque Research and Technology Alliance (BRTA), Donostia-San Sebastián, 20014, Spain
- Neuromuscular Diseases Group, Neurosciences Area, Biodonostia Health Research Institute, Donostia/San Sebastián, 20014, Spain
| | - Alessandro Silvestri
- Center for Cooperative Research in Biomaterials (CIC BiomaGUNE), Basque Research and Technology Alliance (BRTA), Donostia-San Sebastián, 20014, Spain
| | - Haizpea Lasa
- Center for Cooperative Research in Biomaterials (CIC BiomaGUNE), Basque Research and Technology Alliance (BRTA), Donostia-San Sebastián, 20014, Spain
- Neuromuscular Diseases Group, Neurosciences Area, Biodonostia Health Research Institute, Donostia/San Sebastián, 20014, Spain
| | - Donato Mancino
- Center for Cooperative Research in Biomaterials (CIC BiomaGUNE), Basque Research and Technology Alliance (BRTA), Donostia-San Sebastián, 20014, Spain
| | - Maurizio Prato
- Center for Cooperative Research in Biomaterials (CIC BiomaGUNE), Basque Research and Technology Alliance (BRTA), Donostia-San Sebastián, 20014, Spain
- Ikerbasque, Basque Foundation for Science, Bilbao, 48013, Spain
- Department of Chemical and Pharmaceutical Sciences, Universitá Degli Studi di Trieste, Trieste, 34127, Italy
| | - Nuria Alegret
- Center for Cooperative Research in Biomaterials (CIC BiomaGUNE), Basque Research and Technology Alliance (BRTA), Donostia-San Sebastián, 20014, Spain
| |
Collapse
|
7
|
Kwokdinata C, Ramanujam V, Chen J, de Oliveira PN, Nai MH, Chooi WH, Lim CT, Ng SY, David L, Chew SY. Encapsulation of Human Spinal Cord Progenitor Cells in Hyaluronan-Gelatin Hydrogel for Spinal Cord Injury Treatment. ACS APPLIED MATERIALS & INTERFACES 2023; 15:50679-50692. [PMID: 37751213 DOI: 10.1021/acsami.3c07419] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/27/2023]
Abstract
Transplanting human induced pluripotent stem cells (iPSCs)-derived spinal cord progenitor cells (SCPCs) is a promising approach to treat spinal cord injuries. However, stem cell therapies face challenges in cell survival, cell localization to the targeted site, and the control of cell differentiation. Here, we encapsulated SCPCs in thiol-modified hyaluronan-gelatin hydrogels and optimized scaffold mechanical properties and cell encapsulation density to promote cell viability and neuronal differentiation in vitro and in vivo. Different compositions of hyaluronan-gelatin hydrogels formulated by varying concentrations of poly(ethylene glycol) diacrylate were mechanically characterized by using atomic force microscopy. In vitro SCPC encapsulation study showed higher cell viability and proliferation with lower substrate Young's modulus (200 Pa vs 580 Pa) and cell density. Moreover, the soft hydrogels facilitated a higher degree of neuronal differentiation with extended filament structures in contrast to clumped cellular morphologies obtained in stiff hydrogels (p < 0.01). When transplanted in vivo, the optimized SCPC-encapsulated hydrogels resulted in higher cell survival and localization at the transplanted region as compared to cell delivery without hydrogel encapsulation at 2 weeks postimplantation within the rat spinal cord (p < 0.01). Notably, immunostaining demonstrated that the hydrogel-encapsulated SCPCs differentiated along the neuronal and oligodendroglial lineages in vivo. The lack of pluripotency and proliferation also supported the safety of the SCPC transplantation approach. Overall, the injectable hyaluronan-gelatin hydrogel shows promise in supporting the survival and neural differentiation of human SCPCs after transplantation into the spinal cord.
Collapse
Affiliation(s)
- Christy Kwokdinata
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, Singapore 637459, Singapore
| | - Vaibavi Ramanujam
- CNRS@CREATE, Create Tower #08-01, 1 Create Way, Singapore 138602, Singapore
| | - Jiahui Chen
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, Singapore 637459, Singapore
| | | | - Mui Hoon Nai
- Department of Biomedical Engineering, National University of Singapore, Singapore 117576, Singapore
| | - Wai Hon Chooi
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore 138673, Republic of Singapore
| | - Chwee Teck Lim
- Department of Biomedical Engineering, National University of Singapore, Singapore 117576, Singapore
- Mechanobiology Institute, National University of Singapore, Singapore 117411, Singapore
| | - Shi Yan Ng
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore 138673, Republic of Singapore
| | - Laurent David
- CNRS@CREATE, Create Tower #08-01, 1 Create Way, Singapore 138602, Singapore
- Ingénierie des Matériaux Polymères IMP UMR 5223, CNRS, Université Claude Bernard Lyon 1, INSA de Lyon, Université Jean Monnet, Université de Lyon, Villeurbanne F69622, France
| | - Sing Yian Chew
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, Singapore 637459, Singapore
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 308232, Singapore
- School of Materials Science and Engineering, Nanyang Technological University, 50 Nanyang Avenue, Singapore 639798, Singapore
| |
Collapse
|
8
|
Jiao L, Xu T, Du X, Chen X, Jiao Q, Jiang H. The Inhibition Effects of Sodium Nitroprusside on the Survival of Differentiated Neural Stem Cells through the p38 Pathway. Brain Sci 2023; 13:brainsci13030438. [PMID: 36979248 PMCID: PMC10046126 DOI: 10.3390/brainsci13030438] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 02/25/2023] [Accepted: 02/28/2023] [Indexed: 03/08/2023] Open
Abstract
Nitric oxide (NO) is a crucial factor in regulating neuronal development. However, certain effects of NO are complex under different physiological conditions. In this study, we used differentiated neural stem cells (NSCs), which contained neural progenitor cells, neurons, astrocytes, and oligodendrocytes, to observe the physiological effects of sodium nitroprusside (SNP) on the early developmental stage of the nervous system. After SNP treatment for 24 h, the results showed that SNP at 100 μM, 200 μM, 300 μM, and 400 μM concentrations resulted in reduced cell viability and increased cleaved caspase 3 levels, while no significant changes were found at 50 μM. There were no effects on neuronal differentiation in the SNP-treated groups. The phosphorylation of p38 was also significantly upregulated with SNP concentrations of 100 μM, 200 μM, 300 μM, and 400 μM, with no changes for 50 μM concentration in comparison with the control. We also observed that the levels of phosphorylation increased with the increasing concentration of SNP. To further explore the possible role of p38 in SNP-regulated survival of differentiated NSCs, SB202190, the antagonist of p38 mitogen-activated protein kinase, at a concentration of 10 mM, was pretreated for 30 min, and the ratio of phosphorylated p38 was found to be decreased after treatment with SNP. Survival and cell viability increased in the SB202190 and SNP co-treated group. Taken together, our results suggested that p38 is involved in the cell survival of NSCs, regulated by NO.
Collapse
Affiliation(s)
- Lingling Jiao
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, State Key Disciplines: Physiology, Shandong Provincial Collaborative Innovation Center for Neurodegenerative Disorders, School of Basic Medicine, Qingdao University, Qingdao 266071, China
| | - Tongying Xu
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, State Key Disciplines: Physiology, Shandong Provincial Collaborative Innovation Center for Neurodegenerative Disorders, School of Basic Medicine, Qingdao University, Qingdao 266071, China
| | - Xixun Du
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, State Key Disciplines: Physiology, Shandong Provincial Collaborative Innovation Center for Neurodegenerative Disorders, School of Basic Medicine, Qingdao University, Qingdao 266071, China
| | - Xi Chen
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, State Key Disciplines: Physiology, Shandong Provincial Collaborative Innovation Center for Neurodegenerative Disorders, School of Basic Medicine, Qingdao University, Qingdao 266071, China
| | - Qian Jiao
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, State Key Disciplines: Physiology, Shandong Provincial Collaborative Innovation Center for Neurodegenerative Disorders, School of Basic Medicine, Qingdao University, Qingdao 266071, China
- Correspondence:
| | - Hong Jiang
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, State Key Disciplines: Physiology, Shandong Provincial Collaborative Innovation Center for Neurodegenerative Disorders, School of Basic Medicine, Qingdao University, Qingdao 266071, China
- College of Health and Life Science, University of Health and Rehabilitation Sciences, Qingdao 266071, China
| |
Collapse
|
9
|
The Biological Behaviors of Neural Stem Cell Affected by Microenvironment from Host Organotypic Brain Slices under Different Conditions. Int J Mol Sci 2023; 24:ijms24044182. [PMID: 36835592 PMCID: PMC9964775 DOI: 10.3390/ijms24044182] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 02/14/2023] [Accepted: 02/17/2023] [Indexed: 02/22/2023] Open
Abstract
Therapeutic strategies based on neural stem cells (NSCs) transplantation bring new hope for neural degenerative disorders, while the biological behaviors of NSCs after being grafted that were affected by the host tissue are still largely unknown. In this study, we engrafted NSCs that were isolated from a rat embryonic cerebral cortex onto organotypic brain slices to examine the interaction between grafts and the host tissue both in normal and pathological conditions, including oxygen-glucose deprivation (OGD) and traumatic injury. Our data showed that the survival and differentiation of NSCs were strongly influenced by the microenvironment of the host tissue. Enhanced neuronal differentiation was observed in normal conditions, while significantly more glial differentiation was observed in injured brain slices. The process growth of grafted NSCs was guided by the cytoarchitecture of host brain slices and showed the distinct difference between the cerebral cortex, corpus callosum and striatum. These findings provided a powerful resource for unraveling how the host environment determines the fate of grafted NSCs, and raise the prospect of NSCs transplantation therapy for neurological diseases.
Collapse
|
10
|
Integrative Proteome Analysis Revels 3-Hydroxybutyrate Exerts Neuroprotective Effect by Influencing Chromatin Bivalency. Int J Mol Sci 2023; 24:ijms24010868. [PMID: 36614311 PMCID: PMC9821512 DOI: 10.3390/ijms24010868] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 12/13/2022] [Accepted: 12/20/2022] [Indexed: 01/05/2023] Open
Abstract
3-hydroxybutyrate (3OHB) has been proved to act as a neuroprotective molecule in multiple neurodegenerative diseases. Here, we employed a quantitative proteomics approach to assess the changes of the global protein expression pattern of neural cells upon 3OHB administration. In combination with a disease-related, protein-protein interaction network we pinpointed a hub marker, histone lysine 27 trimethylation, which is one of the key epigenetic markers in multiple neurodegenerative diseases. Integrative analysis of transcriptomic and epigenomic datasets highlighted the involvement of bivalent transcription factors in 3OHB-mediated disease protection and its alteration of neuronal development processes. Transcriptomic profiling revealed that 3OHB impaired the fate decision process of neural precursor cells by repressing differentiation and promoting proliferation. Our study provides a new mechanism of 3OHB's neuroprotective effect, in which chromatin bivalency is sensitive to 3OHB alteration and drives its neuroprotective function both in neurodegenerative diseases and in neural development processes.
Collapse
|
11
|
Baek J, Kumar S, Schaffer DV, Im SG. N-Cadherin adhesive ligation regulates mechanosensitive neural stem cell lineage commitment in 3D matrices. Biomater Sci 2022; 10:6768-6777. [PMID: 36314115 PMCID: PMC10195187 DOI: 10.1039/d2bm01349e] [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] [Indexed: 11/21/2022]
Abstract
During differentiation, neural stem cells (NSCs) encounter diverse cues from their niche, including not only biophysical cues from the extracellular matrix (ECM) but also cell-cell communication. However, it is still poorly understood how these cues cumulatively regulate mechanosensitive NSC fate commitment, especially in 3D matrices that better mimic in vivo systems. Here, we develop a click chemistry-based 3D hydrogel material system to fully decouple cell-cell and cell-ECM interactions by functionalizing small peptides: the HAVDI motif from N-cadherin and RGD motif from fibronectin. The hydrogel is engineered to range in stiffness from 75 Pa to 600 Pa. Interestingly, HAVDI-mediated interaction shows increased neurogenesis, except for the softest gel (75 Pa). Moreover, the HAVDI ligation attenuates the mechanosensing state of NSCs, exhibiting restricted cytoskeletal formation and RhoA signaling. Given that mechanosensitive neurogenesis has been reported to be regulated by cytoskeletal formation, our finding suggests that the enhanced neurogenesis in the HAVDI-modified gel may be highly associated with the HAVDI interaction-mediated attenuation of mechanosensing. Furthermore, NSCs in the HAVDI gel shows higher β-catenin activity, which has been known to promote neurogenesis. Our findings provide critical insights into how mechanosensitive NSC fate commitment is regulated as a consequence of diverse interactions in 3D microenvironments.
Collapse
Affiliation(s)
- Jieung Baek
- Dept. of Chemical and Biomolecular Engineering, University of California, Berkeley, Berkeley, CA 94720, USA
- Dept. of Bioengineering, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Sanjay Kumar
- Dept. of Chemical and Biomolecular Engineering, University of California, Berkeley, Berkeley, CA 94720, USA
- Dept. of Bioengineering, University of California, Berkeley, Berkeley, CA 94720, USA
- Dept. of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, CA 94158, USA
| | - David V Schaffer
- Dept. of Chemical and Biomolecular Engineering, University of California, Berkeley, Berkeley, CA 94720, USA
- Dept. of Bioengineering, University of California, Berkeley, Berkeley, CA 94720, USA
- Dept. of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Sung Gap Im
- Dept. of Chemical and Biomolecular Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Republic of Korea.
| |
Collapse
|
12
|
A Tale of Two: When Neural Stem Cells Encounter Hypoxia. Cell Mol Neurobiol 2022:10.1007/s10571-022-01293-6. [DOI: 10.1007/s10571-022-01293-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Accepted: 10/02/2022] [Indexed: 11/12/2022]
|
13
|
An J, Tan R, Hu X, Cai Z, Sun M, Ge Q, Ma W, Li H, Lu H. Kinase inhibit region of SOCS3 attenuates IL6-induced proliferation and astrocytic differentiation of neural stem cells via cross talk between signaling pathways. CNS Neurosci Ther 2022; 29:168-180. [PMID: 36217678 PMCID: PMC9804055 DOI: 10.1111/cns.13992] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 09/07/2022] [Accepted: 09/22/2022] [Indexed: 02/06/2023] Open
Abstract
AIMS Efficiency of neural stem cells (NSCs) therapy for brain injury is restricted by astrogliosis around the damaged region, in which JAK2/STAT3 signaling plays a key role. The SOCS3 that can directly inhibit JAK/STAT3 pathway. Here, we investigated the effects of a fusion peptide that combined kinase inhibitory region (KIR) of SOCS3 and virus trans-activator of transcription (TAT) on biological behavior of cultured NSCs under inflammatory conditions. METHODS NSCs were isolated from embryonic brain of SD rats, TAT-KIR was synthesized, and penetration rate was evaluated by flow cytometry (FACS). CCK8, immunostaining, and FACS were used to detected of TAT-KIR on the proliferation of NSCs. The expressions of GFAP and β tubulin III positive cells induced by IL6 with/without TAT-KIR were examined by immunostaining and Western blotting to observe the NSCs differentiation, and the effect of TAT-KIR on signaling cross talk was observed by Western blotting. RESULTS Penetration rate of TAT-KIR into primary cultured NSCs was up to 94%. TAT-KIR did not affect the growth and viability of NSCs. It significantly reduced the NSCs proliferation that enhanced by IL-6 stimulation via blocking the cell cycle progression from the G0/G1 to S phase. In addition, TAT-KIR attenuated astrocytic differentiation and kept high level of neuronal differentiation derived from IL-6-induced NSCs. The fate of NSCs differentiation under inflammatory conditions was affected by TAT-KIR, which was associated with synchronous inhibition of STAT3 and AKT, while promoting JNK expression. CONCLUSION TAT-KIR mimetic of SOCS3 could be a promising approach for brain repair via regulating the biological behaviors of exogenous NSCs.
Collapse
Affiliation(s)
- Jing An
- Department of Neurobiology, School of Basic Medical SciencesXi'an Jiaotong University Health Science CenterXi'anChina
| | - Ruo‐Lan Tan
- Department of Neurobiology, School of Basic Medical SciencesXi'an Jiaotong University Health Science CenterXi'anChina
| | - Xiao‐Xuan Hu
- Department of Neurobiology, School of Basic Medical SciencesXi'an Jiaotong University Health Science CenterXi'anChina
| | - Zhen‐Lu Cai
- Department of Neurobiology, School of Basic Medical SciencesXi'an Jiaotong University Health Science CenterXi'anChina
| | - Mei‐Qi Sun
- Department of Neurobiology, School of Basic Medical SciencesXi'an Jiaotong University Health Science CenterXi'anChina
| | - Qian Ge
- Department of Neurobiology, School of Basic Medical SciencesXi'an Jiaotong University Health Science CenterXi'anChina
| | - Wen Ma
- Department of Neurobiology, School of Basic Medical SciencesXi'an Jiaotong University Health Science CenterXi'anChina
| | - Hui‐Liang Li
- Faculty of Medical Sciences, Wolfson Institute for Biomedical ResearchUniversity College LondonLondonUK
| | - Hai‐Xia Lu
- Department of Neurobiology, School of Basic Medical SciencesXi'an Jiaotong University Health Science CenterXi'anChina
| |
Collapse
|
14
|
Divergence between Neuronal and Oligodendroglial Cell Fate, in Postnatal Brain Neural Stem Cells, Leads to Divergent Properties in Polymorphic In Vitro Assays. Cells 2022; 11:cells11111743. [PMID: 35681436 PMCID: PMC9179558 DOI: 10.3390/cells11111743] [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: 02/22/2022] [Revised: 05/12/2022] [Accepted: 05/13/2022] [Indexed: 02/01/2023] Open
Abstract
Two main stem cell pools exist in the postnatal mammalian brain that, although they share some “stemness” properties, also exhibit significant differences. Multipotent neural stem cells survive within specialized microenvironments, called niches, and they are vulnerable to ageing. Oligodendroglial lineage-restricted progenitor cells are widely distributed in the brain parenchyma and are more resistant to the effects of ageing. Here, we create polymorphic neural stem cell cultures and allow cells to progress towards the neuronal and the oligodendroglial lineage. We show that the divergence of cell fate is accompanied by a divergence in the properties of progenitors, which reflects their adaptation to life in the niche or the parenchyma. Neurogenesis shows significant spatial restrictions and a dependence on laminin, a major niche component, while oligodendrogenesis shows none of these constraints. Furthermore, the blocking of integrin-β1 leads to opposing effects, reducing neurogenesis and enhancing oligodendrogenesis. Therefore, polymorphic neural stem cell assays can be used to investigate the divergence of postnatal brain stem cells and also to predict the in vivo effects of potential therapeutic molecules targeting stem and progenitor cells, as we do for the microneurotrophin BNN-20.
Collapse
|
15
|
Bierman-Duquette RD, Safarians G, Huang J, Rajput B, Chen JY, Wang ZZ, Seidlits SK. Engineering Tissues of the Central Nervous System: Interfacing Conductive Biomaterials with Neural Stem/Progenitor Cells. Adv Healthc Mater 2022; 11:e2101577. [PMID: 34808031 PMCID: PMC8986557 DOI: 10.1002/adhm.202101577] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 10/31/2021] [Indexed: 12/19/2022]
Abstract
Conductive biomaterials provide an important control for engineering neural tissues, where electrical stimulation can potentially direct neural stem/progenitor cell (NS/PC) maturation into functional neuronal networks. It is anticipated that stem cell-based therapies to repair damaged central nervous system (CNS) tissues and ex vivo, "tissue chip" models of the CNS and its pathologies will each benefit from the development of biocompatible, biodegradable, and conductive biomaterials. Here, technological advances in conductive biomaterials are reviewed over the past two decades that may facilitate the development of engineered tissues with integrated physiological and electrical functionalities. First, one briefly introduces NS/PCs of the CNS. Then, the significance of incorporating microenvironmental cues, to which NS/PCs are naturally programmed to respond, into biomaterial scaffolds is discussed with a focus on electrical cues. Next, practical design considerations for conductive biomaterials are discussed followed by a review of studies evaluating how conductive biomaterials can be engineered to control NS/PC behavior by mimicking specific functionalities in the CNS microenvironment. Finally, steps researchers can take to move NS/PC-interfacing, conductive materials closer to clinical translation are discussed.
Collapse
Affiliation(s)
| | - Gevick Safarians
- Department of Bioengineering, University of California Los Angeles, USA
| | - Joyce Huang
- Department of Bioengineering, University of California Los Angeles, USA
| | - Bushra Rajput
- Department of Bioengineering, University of California Los Angeles, USA
| | - Jessica Y. Chen
- Department of Bioengineering, University of California Los Angeles, USA
- David Geffen School of Medicine, University of California Los Angeles, USA
| | - Ze Zhong Wang
- Department of Bioengineering, University of California Los Angeles, USA
| | | |
Collapse
|
16
|
Song Y, Sharipol A, Uchida H, Ingalls MH, Piraino L, Mereness JA, Moyston T, DeLouise LA, Ovitt CE, Benoit DS. Encapsulation of Primary Salivary Gland Acinar Cell Clusters and Intercalated Ducts (AIDUCs) within Matrix Metalloproteinase (MMP)-Degradable Hydrogels to Maintain Tissue Structure and Function. Adv Healthc Mater 2022; 11:e2101948. [PMID: 34994104 PMCID: PMC8986612 DOI: 10.1002/adhm.202101948] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 12/08/2021] [Indexed: 12/13/2022]
Abstract
Progress in the development of salivary gland regenerative strategies is limited by poor maintenance of the secretory function of salivary gland cells (SGCs) in vitro. To reduce the precipitous loss of secretory function, a modified approach to isolate intact acinar cell clusters and intercalated ducts (AIDUCs), rather than commonly used single cell suspension, is investigated. This isolation approach yields AIDUCs that maintain many of the cell-cell and cell-matrix interactions of intact glands. Encapsulation of AIDUCs in matrix metalloproteinase (MMP)-degradable PEG hydrogels promotes self-assembly into salivary gland mimetics (SGm) with acinar-like structure. Expression of Mist1, a transcription factor associated with secretory function, is detectable throughout the in vitro culture period up to 14 days. Immunohistochemistry also confirms expression of acinar cell markers (NKCC1, PIP and AQP5), duct cell markers (K7 and K5), and myoepithelial cell markers (SMA). Robust carbachol and ATP-stimulated calcium flux is observed within the SGm for up to 14 days after encapsulation, indicating that secretory function is maintained. Though some acinar-to-ductal metaplasia is observed within SGm, it is reduced compared to previous reports. In conclusion, cell-cell interactions maintained within AIDUCs together with the hydrogel microenvironment may be a promising platform for salivary gland regenerative strategies.
Collapse
Affiliation(s)
- Yuanhui Song
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, USA
- Center for Oral Biology, University of Rochester, Rochester, NY, USA
| | - Azmeer Sharipol
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, USA
- Center for Oral Biology, University of Rochester, Rochester, NY, USA
| | - Hitoshi Uchida
- Center for Oral Biology, University of Rochester, Rochester, NY, USA
- Department of Biomedical Genetics, University of Rochester, Rochester, NY, USA
| | - Matthew H. Ingalls
- Center for Oral Biology, University of Rochester, Rochester, NY, USA
- Department of Biomedical Genetics, University of Rochester, Rochester, NY, USA
| | - Lindsay Piraino
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, USA
- Department of Dermatology, University of Rochester, Rochester, NY, USA
| | - Jared A. Mereness
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, USA
- Center for Oral Biology, University of Rochester, Rochester, NY, USA
- Department of Environmental Medicine, University of Rochester, Rochester, NY, USA
| | - Tracey Moyston
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, USA
| | - Lisa A. DeLouise
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, USA
- Department of Dermatology, University of Rochester, Rochester, NY, USA
- Materials Science Program, University of Rochester, Rochester, NY, USA
| | - Catherine E. Ovitt
- Center for Oral Biology, University of Rochester, Rochester, NY, USA
- Department of Biomedical Genetics, University of Rochester, Rochester, NY, USA
- Wilmot Cancer Institute, University of Rochester, Rochester, NY, USA
| | - Danielle S.W. Benoit
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, USA
- Center for Oral Biology, University of Rochester, Rochester, NY, USA
- Department of Biomedical Genetics, University of Rochester, Rochester, NY, USA
- Department of Environmental Medicine, University of Rochester, Rochester, NY, USA
- Wilmot Cancer Institute, University of Rochester, Rochester, NY, USA
- Materials Science Program, University of Rochester, Rochester, NY, USA
- Department of Chemical Engineering, University of Rochester, Rochester, NY, USA
- Center for Musculoskeletal Research, University of Rochester, Rochester, NY, USA
| |
Collapse
|
17
|
Wang F, Wang Q, Wang L, Ren J, Song X, Tian Y, Zheng C, Yang J, Ming D. Low-Intensity Focused Ultrasound Stimulation Ameliorates Working Memory Dysfunctions in Vascular Dementia Rats via Improving Neuronal Environment. Front Aging Neurosci 2022; 14:814560. [PMID: 35264943 PMCID: PMC8899543 DOI: 10.3389/fnagi.2022.814560] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2021] [Accepted: 01/24/2022] [Indexed: 12/19/2022] Open
Abstract
Working memory impairment is one of the remarkable cognitive dysfunctions induced by vascular dementia (VD), and it is necessary to explore an effective treatment. Recently, low-intensity focused ultrasound stimulation (LIFUS) has been found notable neuroprotective effects on some neurological diseases, including VD. However, whether it could ameliorate VD-induced working memory impairment was still not been clarified. The purpose of this study was to address this issue and the underlying mechanism. We established VD rat model using the bilateral common carotid artery occlusion (BCCAO) and applied the LIFUS (center frequency = 0.5 MHz; Ispta = 500 mW/cm2, 10 mins/day) to bilateral medial prefrontal cortex (mPFC) for 2 weeks since 2 weeks after the surgery. The main results showed that the LIFUS could significantly improve the performance of VD rats in the specific working memory tasks (delayed nonmatch-to-sample task and step-down task), which might be associated with the improved synaptic function. We also found the improvement in the cerebral blood flow (CBF) and reduced neuroinflammation in mPFC after LIFUS treatment indicated by the inhibition of Toll-like receptor (TLR4)/nuclear factor kappa B (NF-κB) pathway and the decrease of proinflammatory cytokines. The amelioration of CBF and neuroinflammation may promote the living environment of the neurons in VD which then contribute to the survival of neurons and the improvement in synaptic function. Taken together, our findings indicate that LIFUS targeted mPFC can effectively ameliorate reward-based spatial working memory and fear working memory dysfunctions induced by VD via restoring the living environment, survivability, and synaptic functions of the neurons in mPFC of VD rats. This study adds to the evidence that LIFUS could become a promising and non-invasive treatment strategy for the clinical treatment of central nervous system diseases related to cognitive impairments in the future.
Collapse
Affiliation(s)
- Faqi Wang
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, China
| | - Qian Wang
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, China
| | - Ling Wang
- College of Precision Instruments and Optoelectronics Engineering, Tianjin University, Tianjin, China
- Tianjin Key Laboratory of Brain Science and Neuroengineering, Tianjin, China
| | - Jing Ren
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, China
| | - Xizi Song
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, China
- Tianjin Key Laboratory of Brain Science and Neuroengineering, Tianjin, China
| | - Yutao Tian
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, China
- Tianjin Key Laboratory of Brain Science and Neuroengineering, Tianjin, China
| | - Chenguang Zheng
- College of Precision Instruments and Optoelectronics Engineering, Tianjin University, Tianjin, China
- Tianjin Key Laboratory of Brain Science and Neuroengineering, Tianjin, China
| | - Jiajia Yang
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, China
- College of Precision Instruments and Optoelectronics Engineering, Tianjin University, Tianjin, China
- Tianjin Key Laboratory of Brain Science and Neuroengineering, Tianjin, China
- *Correspondence: Jiajia Yang,
| | - Dong Ming
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, China
- College of Precision Instruments and Optoelectronics Engineering, Tianjin University, Tianjin, China
- Tianjin Key Laboratory of Brain Science and Neuroengineering, Tianjin, China
- Dong Ming,
| |
Collapse
|
18
|
He Z, Ding Y, Mu Y, Xu X, Kong W, Chai R, Chen X. Stem Cell-Based Therapies in Hearing Loss. Front Cell Dev Biol 2021; 9:730042. [PMID: 34746126 PMCID: PMC8567027 DOI: 10.3389/fcell.2021.730042] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Accepted: 10/04/2021] [Indexed: 12/19/2022] Open
Abstract
In recent years, neural stem cell transplantation has received widespread attention as a new treatment method for supplementing specific cells damaged by disease, such as neurodegenerative diseases. A number of studies have proved that the transplantation of neural stem cells in multiple organs has an important therapeutic effect on activation and regeneration of cells, and restore damaged neurons. This article describes the methods for inducing the differentiation of endogenous and exogenous stem cells, the implantation operation and regulation of exogenous stem cells after implanted into the inner ear, and it elaborates the relevant signal pathways of stem cells in the inner ear, as well as the clinical application of various new materials. At present, stem cell therapy still has limitations, but the role of this technology in the treatment of hearing diseases has been widely recognized. With the development of related research, stem cell therapy will play a greater role in the treatment of diseases related to the inner ear.
Collapse
Affiliation(s)
- Zuhong He
- Department of Otorhinolaryngology-Head and Neck Surgery, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Yanyan Ding
- Department of Otorhinolaryngology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yurong Mu
- Department of Otorhinolaryngology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaoxiang Xu
- Department of Otorhinolaryngology-Head and Neck Surgery, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Weijia Kong
- Department of Otorhinolaryngology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Renjie Chai
- State Key Laboratory of Bioelectronics, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, School of Life Sciences and Technology, Southeast University, Nanjing, China.,Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, China.,Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China.,Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, China.,Beijing Key Laboratory of Neural Regeneration and Repair, Capital Medical University, Beijing, China
| | - Xiong Chen
- Department of Otorhinolaryngology-Head and Neck Surgery, Zhongnan Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
19
|
Liu Q, Zhang L, Zhang J. Induced pluripotent stem cell-derived neural progenitor cell transplantation promotes regeneration and functional recovery after post-traumatic stress disorder in rats. Biomed Pharmacother 2021; 133:110981. [PMID: 33186796 DOI: 10.1016/j.biopha.2020.110981] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 11/02/2020] [Accepted: 11/03/2020] [Indexed: 01/09/2023] Open
Abstract
Post-traumatic stress disorder (PTSD) is a mental disorder characterized by hippocampal neuron loss and cognitive dysfunction. The aim of the present study was to investigate the potential functional outcomes of transplantation of induced pluripotent stem cell-derived neural progenitor cells (iPSC-NPCs) for treating PTSD. Human induced pluripotent stem cell (iPSCs), differentiated into neural progenitor cells (NPCs) in vitro, were transplanted into the brain of rat. Following iPSC-NPCs transplantation, cognitive function was determined. The open field test and fear condition test indicated that long-term iPSC-NPCs transplantation ameliorated cognitive dysfunction and reduced freezing time in PTSD rats. Following testing, the brain of rat was analyzed using immunocytochemistry and immunofluorescence. The results revealed that iPSC-NPCs differentiated into neurons replacing the loss of hippocampus neurons, and iPSC-NPCs transplantation showed higher expression of glial fibrillary acidic protein (GFAP) and increased number of NeuN compared with the control group. Moreover, western blot analysis suggested enhanced expression of brain-derived neurotrophic factor (BDNF) in hippocampus tissue of iPSC-NPCs transplanted rats in comparison to the PBS group. Collectively, these findings showed that iPSC-NPCs could promote regeneration and motor function recovery in PTSD model.
Collapse
Affiliation(s)
- Qingzhen Liu
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, No. 163, Xianlin Avenue, Qixia District, Nanjing, 210023, China; Department of Anesthesiology, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, 210002, China
| | - Lidong Zhang
- Department of Anesthesiology, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, 210002, China.
| | - Junfeng Zhang
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, No. 163, Xianlin Avenue, Qixia District, Nanjing, 210023, China.
| |
Collapse
|
20
|
Chen D, Liu J, Wu Z, Li SH. Role of miR-132/methyl-CpG-binding protein 2 in the regulation of neural stem cell differentiation. Neural Regen Res 2021; 16:345-349. [PMID: 32859795 PMCID: PMC7896221 DOI: 10.4103/1673-5374.290908] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Methyl-CpG-binding protein 2 (MeCP2) is a well-known transcription repressor, and mutations in MECP2 cause serious neurological disorders. Many studies have suggested that MeCP2 is involved in neural maturation only, and have not reported its role in neural stem cell differentiation. In the present study, we investigated this possible role of MeCP2 in neural stem cells. We used two different differentiation methods to explore how MeCP2 influences neural stem cell differentiation. When we transfected MeCP2-overexpressing lentivirus into neural stem cells, astrocytic differentiation was impaired. This impaired astrocytic differentiation occurred even in conditions of 20% fetal bovine serum, which favored astrocytic differentiation. In addition, miR-132 had the largest expression change after differentiation among several central nervous system related miRNAs. A luciferase assay confirmed that miR-132 directly targeted MeCP2, and that miR-132 was able to reduce MeCP2 expression at both the RNA and protein levels. The upregulation of miR-132 by miRNA mimics promoted astrocytic differentiation, which was fully recovered by MeCP2 overexpression. These results indicate that miR-132 regulates cell lineage differentiation by reducing MeCP2. The study was approved by the Ethics Committee of Shanghai Tenth People's Hospital of TongJi University, China (approval No. SHDSYY-2018-4748) on March 10, 2018.
Collapse
Affiliation(s)
- Dong Chen
- Department of Orthopedics, Shanghai Tenth People's Hospital, Tongji University, Shanghai, China
| | - Jie Liu
- Department of Orthopedics, Shanghai Tenth People's Hospital, Tongji University, Shanghai, China
| | - Zhong Wu
- Department of Orthopedics, Shanghai Tenth People's Hospital, Tongji University, Shanghai, China
| | - Shao-Hua Li
- Department of Orthopedics, Shanghai Tenth People's Hospital, Tongji University, Shanghai, China
| |
Collapse
|
21
|
Pooyan P, Karamzadeh R, Mirzaei M, Meyfour A, Amirkhan A, Wu Y, Gupta V, Baharvand H, Javan M, Salekdeh GH. The Dynamic Proteome of Oligodendrocyte Lineage Differentiation Features Planar Cell Polarity and Macroautophagy Pathways. Gigascience 2020; 9:giaa116. [PMID: 33128372 PMCID: PMC7601170 DOI: 10.1093/gigascience/giaa116] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Revised: 07/22/2020] [Accepted: 09/28/2020] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Generation of oligodendrocytes is a sophisticated multistep process, the mechanistic underpinnings of which are not fully understood and demand further investigation. To systematically profile proteome dynamics during human embryonic stem cell differentiation into oligodendrocytes, we applied in-depth quantitative proteomics at different developmental stages and monitored changes in protein abundance using a multiplexed tandem mass tag-based proteomics approach. FINDINGS Our proteome data provided a comprehensive protein expression profile that highlighted specific expression clusters based on the protein abundances over the course of human oligodendrocyte lineage differentiation. We identified the eminence of the planar cell polarity signalling and autophagy (particularly macroautophagy) in the progression of oligodendrocyte lineage differentiation-the cooperation of which is assisted by 106 and 77 proteins, respectively, that showed significant expression changes in this differentiation process. Furthermore, differentially expressed protein analysis of the proteome profile of oligodendrocyte lineage cells revealed 378 proteins that were specifically upregulated only in 1 differentiation stage. In addition, comparative pairwise analysis of differentiation stages demonstrated that abundances of 352 proteins differentially changed between consecutive differentiation time points. CONCLUSIONS Our study provides a comprehensive systematic proteomics profile of oligodendrocyte lineage cells that can serve as a resource for identifying novel biomarkers from these cells and for indicating numerous proteins that may contribute to regulating the development of myelinating oligodendrocytes and other cells of oligodendrocyte lineage. We showed the importance of planar cell polarity signalling in oligodendrocyte lineage differentiation and revealed the autophagy-related proteins that participate in oligodendrocyte lineage differentiation.
Collapse
Affiliation(s)
- Paria Pooyan
- Department of Molecular Systems Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, Banihashem St., ACECR, Tehran 16635-148, Iran
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, Banihashem St., ACECR, Tehran 16635-148, Iran
- Department of Brain and Cognitive Science, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, Banihashem St., ACECR, Tehran 16635-148, Iran
| | - Razieh Karamzadeh
- Department of Molecular Systems Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, Banihashem St., ACECR, Tehran 16635-148, Iran
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, Banihashem St., ACECR, Tehran 16635-148, Iran
- Department of Brain and Cognitive Science, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, Banihashem St., ACECR, Tehran 16635-148, Iran
| | - Mehdi Mirzaei
- Department of Molecular Sciences, Macquarie University, North Ryde, Sydney, NSW 2109, Australia
- Australian Proteome Analysis Facility, Macquarie University, North Ryde, NSW 2109, Australia
| | - Anna Meyfour
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Daneshjoo Blv., Velenjak, Tehran 19839-63113, Iran
| | - Ardeshir Amirkhan
- Australian Proteome Analysis Facility, Macquarie University, North Ryde, NSW 2109, Australia
| | - Yunqi Wu
- Australian Proteome Analysis Facility, Macquarie University, North Ryde, NSW 2109, Australia
| | - Vivek Gupta
- Department of Clinical Medicine, Macquarie University, North Ryde, Sydney, NSW 2109, Australia
| | - Hossein Baharvand
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, Banihashem St., ACECR, Tehran 16635-148, Iran
- Department of Brain and Cognitive Science, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, Banihashem St., ACECR, Tehran 16635-148, Iran
- Department of Developmental Biology, University of Science and Culture, Ashrafi Esfahani, Tehran 1461968151, Iran
| | - Mohammad Javan
- Department of Brain and Cognitive Science, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, Banihashem St., ACECR, Tehran 16635-148, Iran
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Jalal AleAhmad, Tehran 14115-111, Iran
| | - Ghasem Hosseini Salekdeh
- Department of Molecular Systems Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, Banihashem St., ACECR, Tehran 16635-148, Iran
- Department of Molecular Sciences, Macquarie University, North Ryde, Sydney, NSW 2109, Australia
| |
Collapse
|
22
|
Yu Y, Li Z, Ma F, Chen Q, Lin L, Xu Q, Li Y, Xin X, Pan P, Huang T, Wang Y, Fei Q, Ge RS. Neurotrophin-3 stimulates stem Leydig cell proliferation during regeneration in rats. J Cell Mol Med 2020; 24:13679-13689. [PMID: 33090725 PMCID: PMC7753877 DOI: 10.1111/jcmm.15886] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Revised: 07/30/2020] [Accepted: 08/17/2020] [Indexed: 11/28/2022] Open
Abstract
Neurotrophin‐3 (NT‐3) acts as an important growth factor to stimulate and control tissue development. The NT‐3 receptor, TRKC, is expressed in rat testis. Its function in regulation of stem Leydig cell development and its underlying mechanism remain unknown. Here, we reported the role of NT‐3 to regulate stem Leydig cell development in vivo and in vitro. Ethane dimethane sulphonate was used to kill all Leydig cells in adult testis, and NT‐3 (10 and 100 ng/testis) was injected intratesticularly from the 14th day after ethane dimethane sulphonate injection for 14 days. NT‐3 significantly reduced serum testosterone levels at doses of 10 and 100 ng/testis without affecting serum luteinizing hormone and follicle‐stimulating hormone levels. NT‐3 increased CYP11A1‐positive Leydig cell number at 100 ng/testis and lowered Leydig cell size and cytoplasmic size at doses of 10 and 100 ng/testis. After adjustment by the Leydig cell number, NT‐3 significantly down‐regulated the expression of Leydig cell genes (Lhcgr, Scarb1, Star, Cyp11a1, Hsd3b1, Cyp17a1, Hsd17b3, Hsd11b1, Insl3, Trkc and Nr5a1) and the proteins. NT‐3 increased the phosphorylation of AKT1 and mTOR, decreased the phosphorylation of 4EBP, thereby increasing ATP5O. In vitro study showed that NT‐3 dose‐dependently stimulated EdU incorporation into stem Leydig cells and inhibited stem Leydig cell differentiation into Leydig cells, thus leading to lower medium testosterone levels and lower expression of Lhcgr, Scarb1, Trkc and Nr5a1 and their protein levels. NT‐3 antagonist Celitinib can antagonize NT‐3 action in vitro. In conclusion, the present study demonstrates that NT‐3 stimulates stem Leydig cell proliferation but blocks the differentiation via TRKC receptor.
Collapse
Affiliation(s)
- Yige Yu
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Zengqiang Li
- Department of Anesthesiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Feifei Ma
- Department of Anesthesiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Quanxu Chen
- Department of Anesthesiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Liben Lin
- Department of Anesthesiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Qiang Xu
- Department of Anesthesiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Yang Li
- Department of Anesthesiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xiu Xin
- Department of Anesthesiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Peipei Pan
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Tongliang Huang
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Yiyan Wang
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Qianjin Fei
- Reproductive Medicine Center, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Ren-Shan Ge
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China.,Department of Anesthesiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
23
|
Connexin 43: A novel ginsenoside Rg1-sensitive target in a rat model of depression. Neuropharmacology 2020; 170:108041. [DOI: 10.1016/j.neuropharm.2020.108041] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2019] [Revised: 03/02/2020] [Accepted: 03/04/2020] [Indexed: 12/31/2022]
|
24
|
Chakraborty A, Brauer S, Diwan A. Possible therapies of Parkinson's disease: A review. J Clin Neurosci 2020; 75:1-4. [PMID: 32247740 DOI: 10.1016/j.jocn.2020.03.024] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Accepted: 03/20/2020] [Indexed: 02/07/2023]
Abstract
Parkinson's disease (PD) is a complex condition with a wide range of symptoms, like impaired movement, tremors, apathy and depression, and many other symptoms. The disease results from degeneration of dopaminergic neural cells. No cure at present but symptomatic some palliative treatments are available to slow down the disease progression. According to the Parkinson's Foundation every year in U.S., approximately 60,000 Americans diagnosed with PD. Nearly one million will be living with PD in the U.S. by 2020, which is more than the combined number of people diagnosed with multiple sclerosis, muscular dystrophy and Amyotrophic Lateral Sclerosis (ALS). There is no diagnostic test for PD, yet, but this article will review all kinds symptomatic and disease-modifying therapy.
Collapse
Affiliation(s)
| | - Sam Brauer
- Allexcel, Inc., 1 Controls Drive, Shelton 06484, CT, United States
| | - Anil Diwan
- Allexcel, Inc., 1 Controls Drive, Shelton 06484, CT, United States
| |
Collapse
|
25
|
Cai Z, Hu X, Tan R, Feng Y, Sun M, Ma N, Li X, Huang L, An J, Ge Q, Lu H. Neuroprotective effect of green tea extractives against oxidative stress by enhancing the survival and proliferation of PC12 cells. Mol Cell Toxicol 2019. [DOI: 10.1007/s13273-019-0042-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
|
26
|
Hong KH, Kim Y, Song S. Fine-Tunable and Injectable 3D Hydrogel for On-Demand Stem Cell Niche. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2019; 6:1900597. [PMID: 31508277 PMCID: PMC6724362 DOI: 10.1002/advs.201900597] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Revised: 05/22/2019] [Indexed: 06/10/2023]
Abstract
Stem-cell-based tissue engineering requires increased stem cell retention, viability, and control of differentiation. The use of biocompatible scaffolds encapsulating stem cells typically addresses the first two problems. To achieve control of stem cell fate, fine-tuned biocompatible scaffolds with bioactive molecules are necessary. However, given that the fine-tuning of stem cell scaffolds is associated with UV irradiation and in situ scaffold gelation, this process is in conflict with injectability. Herein, a fine-tunable and injectable 3D hydrogel system is developed with the use of thermosensitive poly(organophosphazene) bearing β-cyclodextrin (β-CD PPZ) and two types of adamantane-peptides (Ad-peptides) that are associated with mesenchymal stem cell (MSC) differentiation and that serve as stoichiometrically controlled pendants for fine-tuning. Given that complexation of hosts and guests subject to strict stoichiometric control is achieved with simple mixing, these fabricated hydrogels exhibit well-aligned, fine-tuning responses, even in living animals. Injection of MSCs in fine-tuned hydrogels also results in various chondrogenic differentiation levels at three weeks postinjection. This is attributed to the differential controls of Ad-peptides, if MSC preconditioning is excluded. Eventually, the fine-tunable and injectable 3D hydrogel could be applied as platform technology by simply switching the types of peptides bearing adamantane and their stoichiometry.
Collapse
Affiliation(s)
- Ki Hyun Hong
- Center for BiomaterialsBiomedical research InstituteKorea Institute of Science and TechnologySeoul02792Republic of Korea
- Division of Bio‐Medical Science and TechnologyKIST SchoolKorea University of Science and TechnologySeoul02792Republic of Korea
| | - Young‐Min Kim
- Center for BiomaterialsBiomedical research InstituteKorea Institute of Science and TechnologySeoul02792Republic of Korea
| | - Soo‐Chang Song
- Center for BiomaterialsBiomedical research InstituteKorea Institute of Science and TechnologySeoul02792Republic of Korea
- Division of Bio‐Medical Science and TechnologyKIST SchoolKorea University of Science and TechnologySeoul02792Republic of Korea
| |
Collapse
|
27
|
Lee HJ, Ahn SM, Pak ME, Jung DH, Lee SY, Shin HK, Choi BT. Positive effects of α-asarone on transplanted neural progenitor cells in a murine model of ischemic stroke. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2018; 51:151-161. [PMID: 30466612 DOI: 10.1016/j.phymed.2018.09.230] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Revised: 09/20/2018] [Accepted: 09/30/2018] [Indexed: 06/09/2023]
Abstract
BACKGROUND Some traditional Oriental herbal medicines, such as Acorus tatarinowii and Acorus gramineus, produce beneficial effects for cognition enhancement. An active compound in rhizomes and the bark of these plants is α-asarone. PURPOSE This study investigated the effects of α-asarone on the proliferation and differentiation of neural progenitor cells (NPCs) in a primary culture and a murine model of ischemic stroke. METHODS NPCs were isolated from mouse fetal cerebral cortices on embryonic day 15, and all experiments were performed using passage 3 NPCs. We utilized a cell counting kit-8 assay, flow cytometry, western blot, and immunohistochemical analysis to assess proliferation and differentiation of NPCs and employed α-asarone in NPC transplanted ischemic stroke mice to evaluate stroke-related functional recovery using behavioral and immunohistochemical analysis. RESULT Treatment with 1 µM, 3 µM, or 10 μM α-asarone induced significant NPC proliferation compared to vehicle treatment. Induced NPCs expressed the neuronal marker neuronal nuclei (NeuN) or the astrocyte marker S100 calcium-binding protein B (S100β). Both immunohistochemistry and flow cytometry revealed that treatment with α-asarone increased the number of NeuN-immunoreactive cells and decreased the number of S100β-immunoreactive cells. Treatment with α-asarone also increased the expression of β-catenin, cyclin D1, and phosphorylated extracellular signal-regulated kinase (ERK) compared to vehicle treatment. In a murine model of ischemic stroke, treatment with α-asarone and transplanted NPCs alleviated stroke-related functional impairments. The corner and rotarod test results revealed that treatment with α-asarone in the NPC transplanted group had greater-than-additive effects on sensorimotor function and motor balance. Moreover, α-asarone treatment promoted the differentiation of transplanted NPCs into NeuN-, glial fibrillary acidic protein (GFAP)-, platelet-derived growth factor-α (PDGFR-α)-, and 2', 3'-cyclic nucleotide 3'-phosphodiesterase (CNPase)-immunoreactive cells. CONCLUSION α-asarone may promote NPC proliferation and differentiation into neuron-lineage cells by activating β-catenin, cyclin D1, and ERK. Moreover, α-asarone treatment facilitated neurofunctional recovery after NPC transplantation in a murine model of ischemic stroke. Therefore, α-asarone is a potential adjunct treatment to NPC therapy for functional restoration after brain injuries such as ischemic stroke.
Collapse
Affiliation(s)
- Hong Ju Lee
- Department of Korean Medical Science, School of Korean Medicine, Pusan National University, Yangsan 50612, Republic of Korea; Graduate Training Program of Korean Medicine for Healthy-Aging, School of Korean Medicine, Pusan National University, Yangsan 50612, Republic of Korea.
| | - Sung Min Ahn
- Korean Medical Science Research Center for Healthy-Aging, Pusan National University, Yangsan 50612, Republic of Korea.
| | - Malk Eun Pak
- Department of Korean Medical Science, School of Korean Medicine, Pusan National University, Yangsan 50612, Republic of Korea; Graduate Training Program of Korean Medicine for Healthy-Aging, School of Korean Medicine, Pusan National University, Yangsan 50612, Republic of Korea.
| | - Da Hee Jung
- Department of Korean Medical Science, School of Korean Medicine, Pusan National University, Yangsan 50612, Republic of Korea; Graduate Training Program of Korean Medicine for Healthy-Aging, School of Korean Medicine, Pusan National University, Yangsan 50612, Republic of Korea.
| | - Seo-Yeon Lee
- Department of Korean Medical Science, School of Korean Medicine, Pusan National University, Yangsan 50612, Republic of Korea; Graduate Training Program of Korean Medicine for Healthy-Aging, School of Korean Medicine, Pusan National University, Yangsan 50612, Republic of Korea; Korean Medical Science Research Center for Healthy-Aging, Pusan National University, Yangsan 50612, Republic of Korea.
| | - Hwa Kyoung Shin
- Department of Korean Medical Science, School of Korean Medicine, Pusan National University, Yangsan 50612, Republic of Korea; Graduate Training Program of Korean Medicine for Healthy-Aging, School of Korean Medicine, Pusan National University, Yangsan 50612, Republic of Korea; Korean Medical Science Research Center for Healthy-Aging, Pusan National University, Yangsan 50612, Republic of Korea.
| | - Byung Tae Choi
- Department of Korean Medical Science, School of Korean Medicine, Pusan National University, Yangsan 50612, Republic of Korea; Graduate Training Program of Korean Medicine for Healthy-Aging, School of Korean Medicine, Pusan National University, Yangsan 50612, Republic of Korea; Korean Medical Science Research Center for Healthy-Aging, Pusan National University, Yangsan 50612, Republic of Korea.
| |
Collapse
|
28
|
Lin C, Chen P, Chan H, Huang Y, Chang NW. Peroxisome proliferator‐activated receptor alpha accelerates neuronal differentiation and this might involve the mitogen‐activated protein kinase pathway. Int J Dev Neurosci 2018; 71:46-51. [DOI: 10.1016/j.ijdevneu.2018.08.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2018] [Revised: 08/09/2018] [Accepted: 08/21/2018] [Indexed: 01/11/2023] Open
Affiliation(s)
- Chingju Lin
- Department of PhysiologyCollege of Medicine, China Medical UniversityTaichungTaiwan, ROC
| | - Pei‐Yi Chen
- Department of BiochemistryCollege of Medicine, China Medical UniversityTaichungTaiwan, ROC
| | - Hsu‐Chin Chan
- Department of BiochemistryCollege of Medicine, China Medical UniversityTaichungTaiwan, ROC
| | - Yi‐Ping Huang
- Department of PhysiologyCollege of Medicine, China Medical UniversityTaichungTaiwan, ROC
| | - Nai Wen Chang
- Department of BiochemistryCollege of Medicine, China Medical UniversityTaichungTaiwan, ROC
| |
Collapse
|
29
|
Neural stem cell therapies and hypoxic-ischemic brain injury. Prog Neurobiol 2018; 173:1-17. [PMID: 29758244 DOI: 10.1016/j.pneurobio.2018.05.004] [Citation(s) in RCA: 112] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Revised: 03/06/2018] [Accepted: 05/09/2018] [Indexed: 12/13/2022]
Abstract
Hypoxic-ischemic brain injury is a significant cause of morbidity and mortality in the adult as well as in the neonate. Extensive pre-clinical studies have shown promising therapeutic effects of neural stem cell-based treatments for hypoxic-ischemic brain injury. There are two major strategies of neural stem cell-based therapies: transplanting exogenous neural stem cells and boosting self-repair of endogenous neural stem cells. Neural stem cell transplantation has been proved to improve functional recovery after brain injury through multiple by-stander mechanisms (e.g., neuroprotection, immunomodulation), rather than simple cell-replacement. Endogenous neural stem cells reside in certain neurogenic niches of the brain and response to brain injury. Many molecules (e.g., neurotrophic factors) can stimulate or enhance proliferation and differentiation of endogenous neural stem cells after injury. In this review, we first present an overview of neural stem cells during normal brain development and the effect of hypoxic-ischemic injury on the activation and function of endogenous neural stem cells in the brain. We then summarize and discuss the current knowledge of strategies and mechanisms for neural stem cell-based therapies on brain hypoxic-ischemic injury, including neonatal hypoxic-ischemic brain injury and adult ischemic stroke.
Collapse
|