1
|
Miller LN, Walters AE, Denninger JK, Hanson MA, Marshall AH, Johantges AC, Hosawi M, Sebring G, Rieskamp JD, Ding T, Rindani R, Chen KS, Goldberg ME, Senthilvelan S, Volk A, Zhao F, Askwith C, Wester JC, Kirby ED. Neural stem and progenitor cells support and protect adult hippocampal function via vascular endothelial growth factor secretion. Mol Psychiatry 2024:10.1038/s41380-024-02827-8. [PMID: 39528687 DOI: 10.1038/s41380-024-02827-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Revised: 10/31/2024] [Accepted: 11/04/2024] [Indexed: 11/16/2024]
Abstract
Adult neural stem and progenitor cells (NSPCs) reside in the dentate gyrus (DG) of the hippocampus throughout the lifespan of most mammalian species. In addition to generating new neurons, NSPCs may alter their niche via secretion of growth factors and cytokines. We recently showed that adult DG NSPCs secrete vascular endothelial growth factor (VEGF), which is critical for maintaining adult neurogenesis. Here, we asked whether NSPC-derived VEGF alters hippocampal function independent of adult neurogenesis. We found that loss of NSPC-derived VEGF acutely impaired hippocampal memory, caused neuronal hyperexcitability and exacerbated excitotoxic injury. Conversely, we observed that overexpression of VEGF reduced microglial response to excitotoxic injury. We also found that NSPCs generate substantial proportions of total DG VEGF and VEGF disperses widely throughout the DG, both of which help explain how this anatomically-restricted cell population could modulate function broadly. These findings suggest that NSPCs actively support and protect DG function via secreted VEGF, thereby providing a non-neurogenic functional dimension to endogenous NSPCs.
Collapse
Affiliation(s)
- Lisa N Miller
- Department of Psychology, The Ohio State University, Columbus, OH, USA
| | - Ashley E Walters
- Department of Psychology, The Ohio State University, Columbus, OH, USA
| | | | - Meretta A Hanson
- Department of Neuroscience, The Ohio State University, Columbus, OH, USA
| | - Alec H Marshall
- Department of Neuroscience, The Ohio State University, Columbus, OH, USA
| | - Aidan C Johantges
- Department of Neuroscience, The Ohio State University, Columbus, OH, USA
| | - Manal Hosawi
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, USA
| | - Gwendolyn Sebring
- Department of Psychology, The Ohio State University, Columbus, OH, USA
| | - Joshua D Rieskamp
- Department of Psychology, The Ohio State University, Columbus, OH, USA
| | - Tianli Ding
- Department of Psychology, The Ohio State University, Columbus, OH, USA
| | - Raina Rindani
- Department of Psychology, The Ohio State University, Columbus, OH, USA
- UC Health, Cincinnati, OH, USA
| | - Kelly S Chen
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, USA
| | - Megan E Goldberg
- Department of Psychology, The Ohio State University, Columbus, OH, USA
| | | | - Abigail Volk
- Department of Psychology, The Ohio State University, Columbus, OH, USA
| | - Fangli Zhao
- Department of Neuroscience, The Ohio State University, Columbus, OH, USA
| | - Candice Askwith
- Department of Neuroscience, The Ohio State University, Columbus, OH, USA
| | - Jason C Wester
- Department of Neuroscience, The Ohio State University, Columbus, OH, USA
| | - Elizabeth D Kirby
- Department of Psychology, The Ohio State University, Columbus, OH, USA.
- Chronic Brain Injury Center, The Ohio State University, Columbus, OH, USA.
| |
Collapse
|
2
|
Khandia R, Gurjar P, Priyanka, Romashchenko V, Al-Hussain SA, Zaki MEA. Recent advances in stem cell therapy: efficacy, ethics, safety concerns, and future directions focusing on neurodegenerative disorders - a review. Int J Surg 2024; 110:6367-6381. [PMID: 39705668 DOI: 10.1097/js9.0000000000001609] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2024] [Accepted: 04/29/2024] [Indexed: 12/22/2024]
Abstract
Neurodegeneration refers to the gradual loss of neurons and extensive changes in glial cells like tau inclusions in astrocytes and oligodendrocytes, α-synuclein inclusions in oligodendrocytes and SOD1 aggregates in astrocytes along with deterioration in the motor, cognition, learning, and behavior. Common neurodegenerative disorders are Alzheimer's disease (AD), Parkinson's disease (PD), amyotrophic lateral sclerosis (ALS), Huntington's disease (HD), spinocerebellar ataxia (SCA), and supranuclear palsy. There is a lack of effective treatment for neurodegenerative diseases, and scientists are putting their efforts into developing therapies against them. Stem cell therapy has emerged as a hope for neurodegenerative disorders since it is not only the damaged neurons that might be replaced, but other neuromodulators and neuroprotectors are secreted. Stem cell terminal differentiation before implantation ensures the implantation of correct cells and molecular markers like carbonic anhydrase II, CNPase (2',3'-cyclic nucleotide 3'-phosphohydrolase), myelin basic protein (MBP), and myelin oligodendrocyte glycoprotein (MOG) elucidate the differentiation. Secretion of various growth factors like epidermal growth factor (EGF), keratinocyte growth factor (KGF), vascular endothelial growth factor-α (VEGF-α), transforming growth factor (TGF), and macrophage inflammatory protein (MIP) supports cell survival, cell proliferation, blood vessel formation, axon regeneration, and neuroglial functional connection formation at the site of degeneration. Adverse effects of stem cell therapy, like teratogenicity and differentiation in different cells other than the desired one under the influence of microenvironment, are a few key concerns. Post-transplantation improved synaptic plasticity, apoptosis inhibition, and reduction in tau-phosphorylation and amyloid beta (Aβ) production has been observed in Alzheimer's patients. A large number of experimental, preclinical, and clinical studies have been conducted, and encouraging results have been obtained. The present review exhaustively discusses various kinds of stem cells, their usage in treating neurodegenerative disorders, limitations and challenges, and ethical issues related to stem cell therapy.
Collapse
Affiliation(s)
- Rekha Khandia
- Department of Biochemistry and Genetics, Barkatullah University, Bhopal, Madhya Pradesh
| | - Pankaj Gurjar
- Centre for Global Health Research, Saveetha Medical College and Hospital, Saveetha Institute of Medical and Technical Sciences, Chennai, Tamil Nadu
- Department of Science and Engineering, Novel Global Community Educational Foundation, Hebersham, Australia
| | - Priyanka
- Department of Veterinary Microbiology, College of Veterinary Science, Guru AngadDev Veterinary and Animal Sciences University (GADVASU), Rampura Phul, Bathinda, Punjab, India
| | | | - Sami A Al-Hussain
- Department of Chemistry, Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh, Saudi Arabia
| | - Magdi E A Zaki
- Department of Chemistry, Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh, Saudi Arabia
| |
Collapse
|
3
|
Nasrolahi A, Shabani Z, Sadigh-Eteghad S, Salehi-Pourmehr H, Mahmoudi J. Stem Cell Therapy for the Treatment of Parkinson's Disease: What Promise Does it Hold? Curr Stem Cell Res Ther 2024; 19:185-199. [PMID: 36815638 DOI: 10.2174/1574888x18666230222144116] [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: 07/31/2022] [Revised: 01/04/2023] [Accepted: 01/06/2023] [Indexed: 02/24/2023]
Abstract
Parkinson's disease (PD) is a common, progressive neurodegenerative disorder characterized by substantia nigra dopamine cell death and a varied clinical picture that affects older people. Although more than two centuries have passed since the earliest attempts to find a cure for PD, it remains an unresolved problem. With this in mind, cell replacement therapy is a new strategy for treating PD. This novel approach aims to replace degenerated dopaminergic (DAergic) neurons with new ones or provide a new source of cells that can differentiate into DAergic neurons. Induced pluripotent stem cells (iPSCs), mesenchymal stem cells (MSCs), neural stem cells (NSCs), and embryonic stem cells (ESCs) are among the cells considered for transplantation therapies. Recently disease-modifying strategies like cell replacement therapies combined with other therapeutic approaches, such as utilizing natural compounds or biomaterials, are proposed to modify the underlying neurodegeneration. In the present review, we discuss the current advances in cell replacement therapy for PD and summarize the existing experimental and clinical evidence supporting this approach.
Collapse
Affiliation(s)
- Ava Nasrolahi
- Infectious Ophthalmologic Research Center, Health Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
- Cellular and Molecular Research Center, Health Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Zahra Shabani
- Center for Cerebrovascular Research, University of California, San Francisco, California, USA
| | - Saeed Sadigh-Eteghad
- Neurosciences Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hanieh Salehi-Pourmehr
- Research Center for Evidence-Based Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Javad Mahmoudi
- Neurosciences Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
4
|
Jurcau A, Andronie-Cioara FL, Nistor-Cseppento DC, Pascalau N, Rus M, Vasca E, Jurcau MC. The Involvement of Neuroinflammation in the Onset and Progression of Parkinson's Disease. Int J Mol Sci 2023; 24:14582. [PMID: 37834030 PMCID: PMC10573049 DOI: 10.3390/ijms241914582] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Revised: 09/22/2023] [Accepted: 09/24/2023] [Indexed: 10/15/2023] Open
Abstract
Parkinson's disease is a neurodegenerative disease exhibiting the fastest growth in incidence in recent years. As with most neurodegenerative diseases, the pathophysiology is incompletely elucidated, but compelling evidence implicates inflammation, both in the central nervous system and in the periphery, in the initiation and progression of the disease, although it is not yet clear what triggers this inflammatory response and where it begins. Gut dysbiosis seems to be a likely candidate for the initiation of the systemic inflammation. The therapies in current use provide only symptomatic relief, but do not interfere with the disease progression. Nonetheless, animal models have shown promising results with therapies that target various vicious neuroinflammatory cascades. Translating these therapeutic strategies into clinical trials is still in its infancy, and a series of issues, such as the exact timing, identifying biomarkers able to identify Parkinson's disease in early and pre-symptomatic stages, or the proper indications of genetic testing in the population at large, will need to be settled in future guidelines.
Collapse
Affiliation(s)
- Anamaria Jurcau
- Department of Psycho-Neuroscience and Rehabilitation, Faculty of Medicine and Pharmacy, University of Oradea, 410073 Oradea, Romania; (A.J.); (D.C.N.-C.)
| | - Felicia Liana Andronie-Cioara
- Department of Psycho-Neuroscience and Rehabilitation, Faculty of Medicine and Pharmacy, University of Oradea, 410073 Oradea, Romania; (A.J.); (D.C.N.-C.)
| | - Delia Carmen Nistor-Cseppento
- Department of Psycho-Neuroscience and Rehabilitation, Faculty of Medicine and Pharmacy, University of Oradea, 410073 Oradea, Romania; (A.J.); (D.C.N.-C.)
| | - Nicoleta Pascalau
- Department of Psycho-Neuroscience and Rehabilitation, Faculty of Medicine and Pharmacy, University of Oradea, 410073 Oradea, Romania; (A.J.); (D.C.N.-C.)
| | - Marius Rus
- Department of Medical Disciplines, Faculty of Medicine and Pharmacy, University of Oradea, 410087 Oradea, Romania
| | - Elisabeta Vasca
- Department of Oral Rehabilitation, Faculty of Medicine “Vasile Goldis” Arad, 310025 Arad, Romania
| | | |
Collapse
|
5
|
Campero-Romero AN, Real FH, Santana-Martínez RA, Molina-Villa T, Aranda C, Ríos-Castro E, Tovar-Y-Romo LB. Extracellular vesicles from neural progenitor cells promote functional recovery after stroke in mice with pharmacological inhibition of neurogenesis. Cell Death Discov 2023; 9:272. [PMID: 37507361 PMCID: PMC10382527 DOI: 10.1038/s41420-023-01561-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 06/28/2023] [Accepted: 07/14/2023] [Indexed: 07/30/2023] Open
Abstract
Neural progenitor cells (NPCs) of the subventricular zone proliferate in response to ischemic stroke in the adult mouse brain. Newly generated cells have been considered to influence recovery following a stroke. However, the mechanism underlying such protection is a matter of active study since it has been thought that proliferating NPCs mediate their protective effects by secreting soluble factors that promote recovery rather than neuronal replacement in the ischemic penumbra. We tested the hypothesis that this mechanism is mediated by the secretion of multimolecular complexes in extracellular vesicles (EVs). We found that the molecular influence of oxygen and glucose-deprived (OGD) NPCs-derived EVs is very limited in improving overt neurological alterations caused by stroke compared to our recently reported astrocyte-derived EVs. However, when we inhibited the ischemia-triggered proliferation of NPCs with the chronic administration of the DNA synthesis inhibitor Ara-C, the effect of NPC-derived EVs became evident, suggesting that the endogenous protection exerted by the proliferation of NPC is mainly carried out through a mechanism that involves the intercellular communication mediated by EVs. We analyzed the proteomic content of NPC-derived EVs cargo with label-free relative abundance mass spectrometry and identified several molecular mediators of neuronal recovery within these vesicles. Our findings indicate that NPC-derived EVs are protective against the ischemic cascade activated by stroke and, thus, hold significant therapeutic potential.
Collapse
Affiliation(s)
- Aura N Campero-Romero
- Department of Molecular Neuropathology, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| | - Fernando H Real
- Department of Molecular Neuropathology, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| | - Ricardo A Santana-Martínez
- Department of Molecular Neuropathology, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| | - Tonatiuh Molina-Villa
- Department of Cellular and Developmental Biology, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Ciudad de México, México
| | - Cristina Aranda
- Department of Molecular Neuropathology, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| | - Emmanuel Ríos-Castro
- Unidad de Genómica, Proteómica y Metabolómica, LaNSE, Cinvestav-IPN, Ciudad de México, México
| | - Luis B Tovar-Y-Romo
- Department of Molecular Neuropathology, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Ciudad de México, Mexico.
| |
Collapse
|
6
|
Makrygianni EA, Chrousos GP. Neural Progenitor Cells and the Hypothalamus. Cells 2023; 12:1822. [PMID: 37508487 PMCID: PMC10378393 DOI: 10.3390/cells12141822] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 05/22/2023] [Accepted: 06/02/2023] [Indexed: 07/30/2023] Open
Abstract
Neural progenitor cells (NPCs) are multipotent neural stem cells (NSCs) capable of self-renewing and differentiating into neurons, astrocytes and oligodendrocytes. In the postnatal/adult brain, NPCs are primarily located in the subventricular zone (SVZ) of the lateral ventricles (LVs) and subgranular zone (SGZ) of the hippocampal dentate gyrus (DG). There is evidence that NPCs are also present in the postnatal/adult hypothalamus, a highly conserved brain region involved in the regulation of core homeostatic processes, such as feeding, metabolism, reproduction, neuroendocrine integration and autonomic output. In the rodent postnatal/adult hypothalamus, NPCs mainly comprise different subtypes of tanycytes lining the wall of the 3rd ventricle. In the postnatal/adult human hypothalamus, the neurogenic niche is constituted by tanycytes at the floor of the 3rd ventricle, ependymal cells and ribbon cells (showing a gap-and-ribbon organization similar to that in the SVZ), as well as suprachiasmatic cells. We speculate that in the postnatal/adult human hypothalamus, neurogenesis occurs in a highly complex, exquisitely sophisticated neurogenic niche consisting of at least four subniches; this structure has a key role in the regulation of extrahypothalamic neurogenesis, and hypothalamic and extrahypothalamic neural circuits, partly through the release of neurotransmitters, neuropeptides, extracellular vesicles (EVs) and non-coding RNAs (ncRNAs).
Collapse
Affiliation(s)
- Evanthia A Makrygianni
- University Research Institute of Maternal and Child Health & Precision Medicine, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - George P Chrousos
- University Research Institute of Maternal and Child Health & Precision Medicine, National and Kapodistrian University of Athens, 11527 Athens, Greece
| |
Collapse
|
7
|
Attaluri S, Jaimes Gonzalez J, Kirmani M, Vogel AD, Upadhya R, Kodali M, Madhu LN, Rao S, Shuai B, Babu RS, Huard C, Shetty AK. Intranasally administered extracellular vesicles from human induced pluripotent stem cell-derived neural stem cells quickly incorporate into neurons and microglia in 5xFAD mice. Front Aging Neurosci 2023; 15:1200445. [PMID: 37424631 PMCID: PMC10323752 DOI: 10.3389/fnagi.2023.1200445] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Accepted: 06/07/2023] [Indexed: 07/11/2023] Open
Abstract
Introduction Extracellular vesicles (EVs) released by human-induced pluripotent stem cell (hiPSC)-derived neural stem cells (NSCs) have robust antiinflammatory and neurogenic properties due to therapeutic miRNAs and proteins in their cargo. Hence, hiPSC-NSC-EVs are potentially an excellent biologic for treating neurodegenerative disorders, including Alzheimer's disease (AD). Methods This study investigated whether intranasally (IN) administered hiPSC-NSC-EVs would quickly target various neural cell types in the forebrain, midbrain, and hindbrain regions of 3-month-old 5xFAD mice, a model of β-amyloidosis and familial AD. We administered a single dose of 25 × 109 hiPSC-NSC-EVs labeled with PKH26, and different cohorts of naïve and 5xFAD mice receiving EVs were euthanized at 45 min or 6 h post-administration. Results At 45 min post-administration, EVs were found in virtually all subregions of the forebrain, midbrain, and hindbrain of naïve and 5xFAD mice, with predominant targeting and internalization into neurons, interneurons, and microglia, including plaque-associated microglia in 5xFAD mice. EVs also came in contact with the plasma membranes of astrocytic processes and the soma of oligodendrocytes in white matter regions. Evaluation of CD63/CD81 expression with the neuronal marker confirmed that PKH26 + particles found within neurons were IN administered hiPSC-NSC-EVs. At 6 h post-administration, EVs persisted in all cell types in both groups, with the distribution mostly matching what was observed at 45 min post-administration. Area fraction (AF) analysis revealed that, in both naïve and 5xFAD mice, higher fractions of EVs incorporate into forebrain regions at both time points. However, at 45 min post-IN administration, AFs of EVs within cell layers in forebrain regions and within microglia in midbrain and hindbrain regions were lower in 5xFAD mice than naïve mice, implying that amyloidosis reduces EV penetrance. Discussion Collectively, the results provide novel evidence that IN administration of therapeutic hiPSC-NSC-EVs is an efficient avenue for directing such EVs into neurons and glia in all brain regions in the early stage of amyloidosis. As pathological changes in AD are observed in multiple brain areas, the ability to deliver therapeutic EVs into various neural cells in virtually every brain region in the early stage of amyloidosis is attractive for promoting neuroprotective and antiinflammatory effects.
Collapse
|
8
|
Denninger JK, Miller LN, Walters AE, Hosawi M, Sebring G, Rieskamp JD, Ding T, Rindani R, Chen KS, Senthilvelan S, Volk A, Zhao F, Askwith C, Kirby ED. Neural stem and progenitor cells support and protect adult hippocampal function via vascular endothelial growth factor secretion. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.24.537801. [PMID: 37163097 PMCID: PMC10168272 DOI: 10.1101/2023.04.24.537801] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
Adult neural stem and progenitor cells (NSPCs) reside in the dentate gyrus (DG) of the hippocampus throughout the lifespan of most mammalian species. In addition to generating new neurons, NSPCs may alter their niche via secretion of growth factors and cytokines. We recently showed that adult DG NSPCs secrete vascular endothelial growth factor (VEGF), which is critical for maintaining adult neurogenesis. Here, we asked whether NSPC-derived VEGF alters hippocampal function independent of adult neurogenesis. We found that loss of NSPC-derived VEGF acutely impaired hippocampal memory, caused neuronal hyperexcitability and exacerbated excitotoxic injury. We also found that NSPCs generate substantial proportions of total DG VEGF and VEGF disperses broadly throughout the DG, both of which help explain how this anatomically-restricted cell population could modulate function broadly. These findings suggest that NSPCs actively support and protect DG function via secreted VEGF, thereby providing a non-neurogenic functional dimension to endogenous NSPCs.
Collapse
Affiliation(s)
| | - Lisa N. Miller
- Department of Psychology, The Ohio State University, Columbus, OH, USA
| | - Ashley E. Walters
- Department of Psychology, The Ohio State University, Columbus, OH, USA
| | - Manal Hosawi
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, USA
| | - Gwendolyn Sebring
- Department of Psychology, The Ohio State University, Columbus, OH, USA
| | | | - Tianli Ding
- Department of Psychology, The Ohio State University, Columbus, OH, USA
| | - Raina Rindani
- Department of Psychology, The Ohio State University, Columbus, OH, USA
- Current affiliation: UC Health, Cincinnati, OH, USA
| | - Kelly S. Chen
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, USA
| | | | - Abigail Volk
- Department of Psychology, The Ohio State University, Columbus, OH, USA
| | - Fangli Zhao
- Department of Neuroscience, The Ohio State University, Columbus, OH, USA
| | - Candice Askwith
- Department of Neuroscience, The Ohio State University, Columbus, OH, USA
| | - Elizabeth D. Kirby
- Department of Psychology, The Ohio State University, Columbus, OH, USA
- Chronic Brain Injury Center, The Ohio State University, Columbus, OH, USA
| |
Collapse
|
9
|
Umar AK. Stem Cell's Secretome Delivery Systems. Adv Pharm Bull 2023; 13:244-258. [PMID: 37342369 PMCID: PMC10278206 DOI: 10.34172/apb.2023.027] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2021] [Revised: 10/05/2021] [Accepted: 12/31/2021] [Indexed: 09/01/2023] Open
Abstract
Stem cells' secretome contains biomolecules that are ready to give therapeutic activities. However, the biomolecules should not be administered directly because of their in vivo instability. They can be degraded by enzymes or seep into other tissues. There have been some advancements in localized and stabilized secretome delivery systems, which have increased their effectiveness. Fibrous, in situ, or viscoelastic hydrogel, sponge-scaffold, bead powder/ suspension, and bio-mimetic coating can maintain secretome retention in the target tissue and prolong the therapy by sustained release. Porosity, young's modulus, surface charge, interfacial interaction, particle size, adhesiveness, water absorption ability, in situ gel/film, and viscoelasticity of the preparation significantly affect the quality, quantity, and efficacy of the secretome. Therefore, the dosage forms, base materials, and characteristics of each system need to be examined to develop a more optimal secretome delivery system. This article discusses the clinical obstacles and potential solutions for secretome delivery, characterization of delivery systems, and devices used or potentially used in secretome delivery for therapeutic applications. This article concludes that secretome delivery for various organ therapies necessitates the use of different delivery systems and bases. Coating, muco-, and cell-adhesive systems are required for systemic delivery and to prevent metabolism. The lyophilized form is required for inhalational delivery, and the lipophilic system can deliver secretomes across the blood-brain barrier. Nano-sized encapsulation and surface-modified systems can deliver secretome to the liver and kidney. These dosage forms can be administered using devices such as a sprayer, eye drop, inhaler, syringe, and implant to improve their efficacy through dosing, direct delivery to target tissues, preserving stability and sterility, and reducing the immune response.
Collapse
Affiliation(s)
- Abd. Kakhar Umar
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Universitas Padjadjaran, Jatinangor 45363, Indonesia
| |
Collapse
|
10
|
Vaidya M, Sreerama S, Gonzalez-Vega M, Smith J, Field M, Sugaya K. Coculture with Neural Stem Cells May Shift the Transcription Profile of Glioblastoma Multiforme towards Cancer-Specific Stemness. Int J Mol Sci 2023; 24:ijms24043242. [PMID: 36834653 PMCID: PMC9962301 DOI: 10.3390/ijms24043242] [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: 01/04/2023] [Revised: 02/04/2023] [Accepted: 02/06/2023] [Indexed: 02/09/2023] Open
Abstract
Glioblastoma multiforme (GBM) possesses a small but significant population of cancer stem cells (CSCs) thought to play a role in its invasiveness, recurrence, and metastasis. The CSCs display transcriptional profiles for multipotency, self-renewal, tumorigenesis, and therapy resistance. There are two possible theories regarding the origin of CSCs in the context of neural stem cells (NSCs); i.e., NSCs modify cancer cells by conferring them with cancer-specific stemness, or NSCs themselves are transformed into CSCs due to the tumor environment created by cancer cells. To test the theories and to investigate the transcriptional regulation of the genes involved in CSC formation, we cocultured NSC and GBM cell lines together. Where genes related to cancer stemness, drug efflux, and DNA modification were upregulated in GBM, they were downregulated in NSCs upon coculture. These results indicate that cancer cells shift the transcriptional profile towards stemness and drug resistance in the presence of NSCs. Concurrently, GBM triggers NSCs differentiation. Because the cell lines were separated by a membrane (0.4 µm pore size) to prevent direct contact between GBM and NSCs, cell-secreted signaling molecules and extracellular vesicles (EVs) are likely involved in reciprocal communication between NSCs and GBM, causing transcription modification. Understanding the mechanism of CSC creation will aid in the identification of precise molecular targets within the CSCs to exterminate them, which, in turn, will increase the efficacy of chemo-radiation treatment.
Collapse
Affiliation(s)
- Manjusha Vaidya
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL 32816, USA
| | - Sandeep Sreerama
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL 32816, USA
| | - Maxine Gonzalez-Vega
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL 32816, USA
| | - Jonhoi Smith
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL 32816, USA
| | - Melvin Field
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL 32816, USA
- Orlando Neurosurgery, AdventHealth Neuroscience Institute, Orlando, FL 32803, USA
| | - Kiminobu Sugaya
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL 32816, USA
- Correspondence:
| |
Collapse
|
11
|
Birjandi AA, Sharpe P. Potential of extracellular space for tissue regeneration in dentistry. Front Physiol 2022; 13:1034603. [DOI: 10.3389/fphys.2022.1034603] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Accepted: 10/24/2022] [Indexed: 11/19/2022] Open
Abstract
With the proven relationship between oral and general health and the growing aging population, it is pivotal to provide accessible therapeutic approaches to regenerate oral tissues and restore clinical function. However, despite sharing many core concepts with medicine, dentistry has fallen behind the progress in precision medicine and regenerative treatments. Stem cell therapies are a promising avenue for tissue regeneration, however, ethical, safety and cost issues may limit their clinical use. With the significance of paracrine signalling in stem cell and tissue regeneration, extracellular space comprising of the cell secretome, and the extracellular matrix can serve as a potent source for tissue regeneration. Extravesicles are secreted and naturally occurring vesicles with biologically active cargo that can be harvested from the extracellular space. These vesicles have shown great potential as disease biomarkers and can be used in regenerative medicine. As a cell free therapy, secretome and extracellular vesicles can be stored and transferred easily and pose less ethical and safety risks in clinical application. Since there are currently many reviews on the secretome and the biogenesis, characterization and function of extracellular vesicles, here we look at the therapeutic potential of extracellular space to drive oral tissue regeneration and the current state of the field in comparison to regenerative medicine.
Collapse
|
12
|
The neural stem cell secretome across neurodevelopment. Exp Neurol 2022; 355:114142. [PMID: 35709983 DOI: 10.1016/j.expneurol.2022.114142] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 06/03/2022] [Accepted: 06/07/2022] [Indexed: 11/21/2022]
Abstract
Neural stem cell (NSC) based therapies are at the forefront of regenerative medicine strategies to combat illness and injury of the central nervous system (CNS). In addition to their ability to produce new cells, NSCs secrete a variety of products, known collectively as the NSC secretome, that have been shown to ameliorate CNS disease pathology and promote recovery. As pre-clinical and clinical research to harness the NSC secretome for therapeutic purposes advances, a more thorough understanding of the endogenous NSC secretome can provide useful insight into the functional capabilities of NSCs. In this review, we focus on research investigating the autocrine and paracrine functions of the endogenous NSC secretome across life. Throughout development and adulthood, we find evidence that the NSC secretome is a critical component of how endogenous NSCs regulate themselves and their niche. We also find gaps in current literature, most notably in the clinically-relevant domain of endogenous NSC paracrine function in the injured CNS. Future investigations to further define the endogenous NSC secretome and its role in CNS tissue regulation are necessary to bolster our understanding of NSC-niche interactions and to aid in the generation of safe and effective NSC-based therapies.
Collapse
|
13
|
Willis CM, Nicaise AM, Krzak G, Ionescu RB, Pappa V, D'Angelo A, Agarwal R, Repollés-de-Dalmau M, Peruzzotti-Jametti L, Pluchino S. Soluble factors influencing the neural stem cell niche in brain physiology, inflammation, and aging. Exp Neurol 2022; 355:114124. [DOI: 10.1016/j.expneurol.2022.114124] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Revised: 05/16/2022] [Accepted: 05/21/2022] [Indexed: 11/27/2022]
|
14
|
Lee EJ, Choi Y, Lee HJ, Hwang DW, Lee DS. Human neural stem cell-derived extracellular vesicles protect against Parkinson's disease pathologies. J Nanobiotechnology 2022; 20:198. [PMID: 35468855 PMCID: PMC9040239 DOI: 10.1186/s12951-022-01356-2] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 03/06/2022] [Indexed: 02/06/2023] Open
Abstract
Background Neural stem cells (NSCs) have the ability to generate a variety of functional neural cell types and have a high potential for neuronal cell regeneration and recovery. Thus, they been recognized as the best source of cell therapy for neurodegenerative diseases, such as Parkinson’s disease (PD). Owing to the possibility of paracrine effect-based therapeutic mechanisms and easier clinical accessibility, extracellular vesicles (EVs), which possess very similar bio-functional components from their cellular origin, have emerged as potential alternatives in regenerative medicine. Material and methods EVs were isolated from human fibroblast (HFF) and human NSC (F3 cells). The supernatant of the cells was concentrated by a tangential flow filtration (TFF) system. Then, the final EVs were isolated using a total EV isolation kit. Results In this study, we demonstrate the potential protective effect of human NSC-derived EVs, showing the prevention of PD pathologies in 6-hydroxydopamine (6-OHDA)-induced in vitro and in vivo mouse models. Human NSC and F3 cell (F3)-derived EVs reduced the intracellular reactive oxygen species (ROS) and associated apoptotic pathways. In addition, F3-derived EVs induced downregulation of pro-inflammatory factors and significantly decreased 6-OHDA-induced dopaminergic neuronal loss in vivo. F3 specific microRNAs (miRNAs) such as hsa-mir-182-5p, hsa-mir-183-5p, hsa-mir-9, and hsa-let-7, which are involved in cell differentiation, neurotrophic function, and immune modulation, were found in F3-derived EVs. Conclusions We report that human NSC-derived EVs show an effective neuroprotective property in an in vitro transwell system and in a PD model. The EVs clearly decreased ROS and pro-inflammatory cytokines. Taken together, these results indicate that NSC-derived EVs could potentially help prevent the neuropathology and progression of PD. Graphical Abstract ![]()
Supplementary Information The online version contains supplementary material available at 10.1186/s12951-022-01356-2.
Collapse
Affiliation(s)
- Eun Ji Lee
- Department of Nuclear Medicine, Seoul National University College of Medicine, 101 Daehak-ro, Jongno-gu, Seoul, 03080, South Korea.,Department of Nuclear Medicine, Seoul National University Hospital, Seoul, South Korea.,Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, South Korea
| | - Yoori Choi
- Department of Nuclear Medicine, Seoul National University College of Medicine, 101 Daehak-ro, Jongno-gu, Seoul, 03080, South Korea.,Department of Nuclear Medicine, Seoul National University Hospital, Seoul, South Korea.,Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, South Korea
| | - Hong J Lee
- College of Medicine and Medical Research Institute, Chungbuk National University, Cheongju, South Korea.,Research Institute, huMetaCELL Inc., 220, Bugwang-ro, Puchon, Gyeonggi-do, Republic of Korea
| | - Do Won Hwang
- Department of Nuclear Medicine, Seoul National University College of Medicine, 101 Daehak-ro, Jongno-gu, Seoul, 03080, South Korea. .,THERABEST, Inc., Seocho-daero 40-gil 41, Seoul, 06656, South Korea.
| | - Dong Soo Lee
- Department of Nuclear Medicine, Seoul National University College of Medicine, 101 Daehak-ro, Jongno-gu, Seoul, 03080, South Korea. .,Department of Nuclear Medicine, Seoul National University Hospital, Seoul, South Korea. .,Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, South Korea.
| |
Collapse
|
15
|
Pedachenko EG, Liubich LD, Staino LP, Egorova DM, Skaterna TD. Neuroregenerative “Bystander”-Effects of Conditioned Media from Adipose Tissue-Derived Fibroblast-Like Cells in Vitro. CYTOL GENET+ 2022. [DOI: 10.3103/s0095452722020098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
16
|
Emerging Roles for the Orphan GPCRs, GPR37 and GPR37 L1, in Stroke Pathophysiology. Int J Mol Sci 2022; 23:ijms23074028. [PMID: 35409385 PMCID: PMC9000135 DOI: 10.3390/ijms23074028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 03/31/2022] [Accepted: 04/02/2022] [Indexed: 11/17/2022] Open
Abstract
Recent studies have shed light on the diverse and complex roles of G-protein coupled receptors (GPCRs) in the pathophysiology of stroke. These receptors constitute a large family of seven transmembrane-spanning proteins that play an intricate role in cellular communication mechanisms which drive both tissue injury and repair following ischemic stroke. Orphan GPCRs represent a unique sub-class of GPCRs for which no natural ligands have been found. Interestingly, the majority of these receptors are expressed within the central nervous system where they represent a largely untapped resource for the treatment of neurological diseases. The focus of this review will thus be on the emerging roles of two brain-expressed orphan GPCRs, GPR37 and GPR37 L1, in regulating various cellular and molecular processes underlying ischemic stroke.
Collapse
|
17
|
Kumar A, Yun H, Funderburgh ML, Du Y. Regenerative therapy for the Cornea. Prog Retin Eye Res 2022; 87:101011. [PMID: 34530154 PMCID: PMC8918435 DOI: 10.1016/j.preteyeres.2021.101011] [Citation(s) in RCA: 57] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 09/08/2021] [Accepted: 09/09/2021] [Indexed: 12/13/2022]
Abstract
The cornea is the outmost layer of the eye, unique in its transparency and strength. The cornea not only transmits the light essential for vision, also refracts light, giving focus to images. Each of the three layers of the cornea has properties essential for the function of vision. Although the epithelium can often recover from injury quickly by cell division, loss of limbal stem cells can cause severe corneal surface abnormalities leading to corneal blindness. Disruption of the stromal extracellular matrix and loss of cells determining this structure, the keratocytes, leads to corneal opacity. Corneal endothelium is the inner part of the cornea without self-renewal capacity. It is very important to maintain corneal dehydration and transparency. Permanent damage to the corneal stroma or endothelium can be effectively treated by corneal transplantation; however, there are drawbacks to this procedure, including a shortage of donors, the need for continuing treatment to prevent rejection, and limits to the survival of the graft, averaging 10-20 years. There exists a need for new strategies to promote regeneration of the stromal structure and restore vision. This review highlights critical contributions in regenerative medicine with the aim of corneal reconstruction after injury or disease. These approaches include corneal stromal stem cells, corneal limbal stem cells, embryonic stem cells, and other adult stem cells, as well as induced pluripotent stem cells. Stem cell-derived trophic factors in the forms of secretomes or exosomes for corneal regeneration are also discussed. Corneal sensory nerve regeneration promoting corneal transparency is discussed. This article provides description of the up-to-date options for corneal regeneration and presents exciting possible avenues for future studies toward clinical applications for corneal regeneration.
Collapse
Affiliation(s)
- Ajay Kumar
- Department of Ophthalmology, University of Pittsburgh, Pittsburgh, PA 15213
| | - Hongmin Yun
- Department of Ophthalmology, University of Pittsburgh, Pittsburgh, PA 15213
| | | | - Yiqin Du
- Department of Ophthalmology, University of Pittsburgh, Pittsburgh, PA, 15213, USA; Department of Developmental Biology, University of Pittsburgh, Pittsburgh, PA, 15213, USA; McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, 15213, USA.
| |
Collapse
|
18
|
Zhang J, Hu Q, Jiang X, Wang S, Zhou X, Lu Y, Huang X, Duan H, Zhang T, Ge H, Yu A. Actin Alpha 2 Downregulation Inhibits Neural Stem Cell Proliferation and Differentiation into Neurons through Canonical Wnt/ β-Catenin Signaling Pathway. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:7486726. [PMID: 35186189 PMCID: PMC8850075 DOI: 10.1155/2022/7486726] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Revised: 12/27/2021] [Accepted: 01/05/2022] [Indexed: 11/29/2022]
Abstract
Our previous study has shown that actin alpha 2 (ACTA2) is expressed in NSC and ACTA2 downregulation inhibits NSC migration by increasing RhoA expression and decreasing the expression of Rac1 to curb actin filament polymerization. Given that proliferation and differentiation are the two main characteristics of NSC, the role of ACTA2 downregulation in the proliferation and differentiation of NSC remains elusive. Here, the results demonstrated that ACTA2 downregulation using ACTA2 siRNA held the potential of inhibiting NSC proliferation using cell counting kit-8 (CCK8) and immunostaining. Then, our data illustrated that ACTA2 downregulation attenuated NSC differentiation into neurons, while directing NSC into astrocytes and oligodendrocytes using immunostaining and immunoblotting. Thereafter, the results revealed that the canonical Wnt/β-catenin pathway was involved in the effect of ACTA2 downregulation on the proliferation and differentiation of NSC through upregulating p-β-catenin and decreasing β-catenin due to inactivating GSK-3β, while this effect could be partially abolished with administration of CHIR99012, a GSK-3 inhibitor. Collectively, these results indicate that ACTA2 downregulation inhibits NSC proliferation and differentiation into neurons through inactivation of the canonical Wnt/β-catenin pathway. The aim of the present study is to elucidate the role of ACTA2 in proliferation and differentiation of NSC and to provide an intervention target for promoting NSC proliferation and properly directing NSC differentiation.
Collapse
Affiliation(s)
- Ji Zhang
- Department of Emergency, Affiliated Hospital of Zunyi Medical University, 563003 Zunyi, Guizhou, China
| | - Quan Hu
- Department of Emergency, Affiliated Hospital of Zunyi Medical University, 563003 Zunyi, Guizhou, China
| | - Xuheng Jiang
- Department of Emergency, Affiliated Hospital of Zunyi Medical University, 563003 Zunyi, Guizhou, China
| | - Shuhong Wang
- Department of Emergency, Affiliated Hospital of Zunyi Medical University, 563003 Zunyi, Guizhou, China
| | - Xin Zhou
- Department of Emergency, Affiliated Hospital of Zunyi Medical University, 563003 Zunyi, Guizhou, China
| | - Yuanlan Lu
- Department of Emergency, Affiliated Hospital of Zunyi Medical University, 563003 Zunyi, Guizhou, China
| | - Xiaofei Huang
- Department of Emergency, Affiliated Hospital of Zunyi Medical University, 563003 Zunyi, Guizhou, China
| | - Haizhen Duan
- Department of Emergency, Affiliated Hospital of Zunyi Medical University, 563003 Zunyi, Guizhou, China
| | - Tianxi Zhang
- Department of Emergency, Affiliated Hospital of Zunyi Medical University, 563003 Zunyi, Guizhou, China
| | - Hongfei Ge
- Department of Emergency, Affiliated Hospital of Zunyi Medical University, 563003 Zunyi, Guizhou, China
| | - Anyong Yu
- Department of Emergency, Affiliated Hospital of Zunyi Medical University, 563003 Zunyi, Guizhou, China
| |
Collapse
|
19
|
Improved Survival and Regeneration of Irradiated Mouse Neural Stem Cells after Co-Culturing with Non-Irradiated Mouse Neural Stem Cells or Mesenchymal Stem Cells from the Adipose Tissue. Bull Exp Biol Med 2021; 172:228-235. [PMID: 34855083 DOI: 10.1007/s10517-021-05368-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Indexed: 01/01/2023]
Abstract
We studied the effect of neural stem cells (NSC) and mesenchymal stem cells (MSC) from mouse adipose tissue on survival, clonogenic activity, and senescence of NSC after exposure to γ-radiation. It was found that survival and clonogenic activity of NSC irradiated in doses of 1 and 2 Gy was enhanced when irradiated cells were co-cultured with non-irradiated NSC and MSC in permeable Transwell inserts. The proportion of senescent NSC (cells with high β-galactosidase activity) increased with increasing irradiation dose. Co-culturing with non-irradiated NSC in 3 days after irradiation in a dose of 1 Gy led to a decrease in the proportion of senescent cells among irradiated NSC. Factors secreted by NSC and MSC can become the basis for the development of means for prevention and treatment of damage to brain cells resulting from radiation therapy of head and neck cancer.
Collapse
|
20
|
Silva RC, Domingues HS, Salgado AJ, Teixeira FG. From regenerative strategies to pharmacological approaches: can we fine-tune treatment for Parkinson's disease? Neural Regen Res 2021; 17:933-936. [PMID: 34558504 PMCID: PMC8552835 DOI: 10.4103/1673-5374.324827] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Parkinson's disease is the second most prevalent neurodegenerative disorder worldwide. Clinically, it is characterized by severe motor complications caused by progressive degeneration of dopaminergic neurons. Current treatment is focused on mitigating the symptoms through the administration of levodopa, rather than on preventing dopaminergic neuronal damage. Therefore, the use and development of neuroprotective/disease-modifying strategies is an absolute need that can lead to promising gains on translational research of Parkinson's disease. For instance, N-acetylcysteine, a natural compound with strong antioxidant effects, has been shown to modulate oxidative stress, preventing dopamine-induced cell death. Despite the evidence of neuroprotective and modulatory effects of this drug, as far as we know, it does not induce per se any regenerative process. Therefore, it would be of interest to combine the latter with innovative therapies that induce dopaminergic neurons repair or even differentiation, as stem cell-based strategies. Stem cells secretome has been proposed as a promising therapeutic approach for Parkinson's disease, given its ability to modulate cell viability/preservation of dopaminergic neurons. Such approach represents a shift in the paradigm, showing that cell-transplantation free therapies based on the use of stem cells secretome may represent a potential alternative for regenerative medicine of Parkinson's disease. Thus, in this review, we address the current understanding of the potential combination of stem cell free-based strategies and neuroprotective/disease-modifying strategies as a new paradigm for the treatment of central nervous system neurodegenerative diseases, like Parkinson's disease.
Collapse
Affiliation(s)
- Rita Caridade Silva
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga; ICVS/3B's Associate Lab, PT Government Associated Lab, Braga/Guimarães, Portugal
| | - Helena Sofia Domingues
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga; ICVS/3B's Associate Lab, PT Government Associated Lab, Braga/Guimarães, Portugal
| | - António J Salgado
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga; ICVS/3B's Associate Lab, PT Government Associated Lab, Braga/Guimarães, Portugal
| | - Fábio G Teixeira
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga; ICVS/3B's Associate Lab, PT Government Associated Lab, Braga/Guimarães, Portugal
| |
Collapse
|
21
|
Schnatz A, Müller C, Brahmer A, Krämer‐Albers E. Extracellular Vesicles in neural cell interaction and CNS homeostasis. FASEB Bioadv 2021; 3:577-592. [PMID: 34377954 PMCID: PMC8332475 DOI: 10.1096/fba.2021-00035] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2021] [Revised: 04/16/2021] [Accepted: 04/22/2021] [Indexed: 12/15/2022] Open
Abstract
Central nervous system (CNS) homeostasis critically depends on the interaction between neurons and glia cells. Extracellular vesicles (EVs) recently emerged as versatile messengers in CNS cell communication. EVs are released by neurons and glia in activity-dependent manner and address multiple target cells within and outside the nervous system. Here, we summarize the recent advances in understanding the physiological roles of EVs in the nervous system and their ability to deliver signals across the CNS barriers. In addition to the disposal of cellular components via EVs and clearance by phagocytic cells, EVs are involved in plasticity-associated processes, mediate trophic support and neuroprotection, promote axonal maintenance, and modulate neuroinflammation. While individual functional components of the EV cargo are becoming progressively identified, the role of neural EVs as compound multimodal signaling entities remains to be elucidated. Novel transgenic models and imaging technologies allow EV tracking in vivo and provide further insight into EV targeting and their mode of action. Overall, EVs represent key players in the maintenance of CNS homeostasis essential for the lifelong performance of neural networks and thus provide a wide spectrum of biomedical applications.
Collapse
Affiliation(s)
- Andrea Schnatz
- Institute of Developmental Biology and NeurobiologyBiology of Extracellular VesiclesUniversity of MainzMainzGermany
| | - Christina Müller
- Institute of Developmental Biology and NeurobiologyBiology of Extracellular VesiclesUniversity of MainzMainzGermany
| | - Alexandra Brahmer
- Institute of Developmental Biology and NeurobiologyBiology of Extracellular VesiclesUniversity of MainzMainzGermany
| | - Eva‐Maria Krämer‐Albers
- Institute of Developmental Biology and NeurobiologyBiology of Extracellular VesiclesUniversity of MainzMainzGermany
| |
Collapse
|
22
|
Owino S, Giddens MM, Jiang JG, Nguyen TT, Shiu FH, Lala T, Gearing M, McCrary MR, Gu X, Wei L, Yu SP, Hall RA. GPR37 modulates progenitor cell dynamics in a mouse model of ischemic stroke. Exp Neurol 2021; 342:113719. [PMID: 33839144 PMCID: PMC9826632 DOI: 10.1016/j.expneurol.2021.113719] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 03/27/2021] [Accepted: 04/06/2021] [Indexed: 01/11/2023]
Abstract
The generation of neural stem and progenitor cells following injury is critical for the function of the central nervous system, but the molecular mechanisms modulating this response remain largely unknown. We have previously identified the G protein-coupled receptor 37 (GPR37) as a modulator of ischemic damage in a mouse model of stroke. Here we demonstrate that GPR37 functions as a critical negative regulator of progenitor cell dynamics and gliosis following ischemic injury. In the central nervous system, GPR37 is enriched in mature oligodendrocytes, but following injury we have found that its expression is dramatically increased within a population of Sox2-positive progenitor cells. Moreover, the genetic deletion of GPR37 did not alter the number of mature oligodendrocytes following injury but did markedly increase the number of both progenitor cells and injury-induced Olig2-expressing glia. Alterations in the glial environment were further evidenced by the decreased activation of oligodendrocyte precursor cells. These data reveal that GPR37 regulates the response of progenitor cells to ischemic injury and provides new perspectives into the potential for manipulating endogenous progenitor cells following stroke.
Collapse
Affiliation(s)
- Sharon Owino
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Michelle M. Giddens
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Jessie G. Jiang
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - TrangKimberly T. Nguyen
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Fu Hung Shiu
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Trisha Lala
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Marla Gearing
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA;,Department of Neurology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Myles R. McCrary
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Xiaohuan Gu
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Ling Wei
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Shan P. Yu
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA 30322, USA;,Center for Visual and Neurocognitive Rehabilitation, Atlanta Veterans Affair Medical Center, Decatur, GA 30033, USA
| | - Randy A. Hall
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA 30322, USA
| |
Collapse
|
23
|
Richard SA, Sackey M. Elucidating the Pivotal Neuroimmunomodulation of Stem Cells in Spinal Cord Injury Repair. Stem Cells Int 2021; 2021:9230866. [PMID: 34341666 PMCID: PMC8325586 DOI: 10.1155/2021/9230866] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2021] [Revised: 07/03/2021] [Accepted: 07/17/2021] [Indexed: 12/11/2022] Open
Abstract
Spinal cord injury (SCI) is a distressing incident with abrupt onset of the motor as well as sensory dysfunction, and most often, the injury occurs as result of high-energy or velocity accidents as well as contact sports and falls in the elderly. The key challenges associated with nerve repair are the lack of self-repair as well as neurotrophic factors and primary and secondary neuronal apoptosis, as well as factors that prevent the regeneration of axons locally. Neurons that survive the initial traumatic damage may be lost due to pathogenic activities like neuroinflammation and apoptosis. Implanted stem cells are capable of differentiating into neural cells that replace injured cells as well as offer local neurotrophic factors that aid neuroprotection, immunomodulation, axonal sprouting, axonal regeneration, and remyelination. At the microenvironment of SCI, stem cells are capable of producing growth factors like brain-derived neurotrophic factor and nerve growth factor which triggers neuronal survival as well as axonal regrowth. Although stem cells have proven to be of therapeutic value in SCI, the major disadvantage of some of the cell types is the risk for tumorigenicity due to the contamination of undifferentiated cells prior to transplantation. Local administration of stem cells via either direct cellular injection into the spinal cord parenchyma or intrathecal administration into the subarachnoid space is currently the best transplantation modality for stem cells during SCI.
Collapse
Affiliation(s)
- Seidu A. Richard
- Department of Medicine, Princefield University, P.O. Box MA128, Ho, Ghana
| | - Marian Sackey
- Department of Pharmacy, Ho Teaching Hospital, P.O. Box MA-374, Ho, Ghana
| |
Collapse
|