1
|
Radoszkiewicz K, Bzinkowska A, Chodkowska M, Rybkowska P, Sypecka M, Zembrzuska-Kaska I, Sarnowska A. Deciphering the impact of cerebrospinal fluid on stem cell fate as a new mechanism to enhance clinical therapy development. Front Neurosci 2024; 17:1332751. [PMID: 38282622 PMCID: PMC10811009 DOI: 10.3389/fnins.2023.1332751] [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: 11/03/2023] [Accepted: 12/29/2023] [Indexed: 01/30/2024] Open
Abstract
Neural stem cells (NSCs) hold a very significant promise as candidates for cell therapy due to their robust neuroprotective and regenerative properties. Preclinical studies using NSCs have shown enough encouraging results to perform deeper investigations into more potential clinical applications. Nevertheless, our knowledge regarding neurogenesis and its underlying mechanisms remains incomplete. To understand them better, it seems necessary to characterize all components of neural stem cell niche and discover their role in physiology and pathology. Using NSCs in vivo brings challenges including limited cell survival and still inadequate integration within host tissue. Identifying overlooked factors that might influence these outcomes becomes pivotal. In this review, we take a deeper examination of the influence of a fundamental element that is present in the brain, the cerebrospinal fluid (CSF), which still remains relatively unexplored. Its role in neurogenesis could be instrumental to help find novel therapeutic solutions for neurological disorders, eventually advancing our knowledge on central nervous system (CNS) regeneration and repair.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Anna Sarnowska
- Translational Platform for Regenerative Medicine, Mossakowski Medical Research Institute, Polish Academy of Sciences, Warsaw, Poland
| |
Collapse
|
2
|
Mallardo M, Signoriello E, Lus G, Daniele A, Nigro E. Adiponectin Alleviates Cell Injury due to Cerebrospinal Fluid from Multiple Sclerosis Patients by Inhibiting Oxidative Stress and Proinflammatory Response. Biomedicines 2023; 11:1692. [PMID: 37371787 DOI: 10.3390/biomedicines11061692] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Revised: 06/05/2023] [Accepted: 06/08/2023] [Indexed: 06/29/2023] Open
Abstract
Multiple sclerosis (MS) is the most common disabling neurological disease characterized by chronic inflammation and neuronal cell viability impairment. Based on previous studies reporting that adiponectin exhibits neuroprotective effects in some models of neurodegenerative diseases, we analyzed the effects of AdipoRon treatment, alone or in combination with the cerebrospinal fluid of patients with MS (MS-CSF), to verify whether this adipokine acts on the basal neuronal cellular processes. To this aim, SH-SY5Y and U-87 cells (models of neuronal and glial cells, respectively) were exposed to MS-CSF alone or in co-treatment with AdipoRon. The cell viability was determined via MTT assay, and the possible underlying mechanisms were investigated via the alterations of oxidative stress and inflammation. MTT assay confirmed that AdipoRon alone did not affect the viability of both cell lines; whereas, when used in combination with MS-CSF, it reduces MS-CSF inhibitory effects on the viability of both SH-SY5Y and U-87 cell lines. In addition, MS-CSF treatment causes an increase in pro-inflammatory cytokines, whereas it determines the reduction in anti-inflammatory IL-10. Interestingly, the co-administration of AdipoRon counteracts the MS-CSF-induced production of pro-inflammatory cytokines, whereas it determines an enhancement of IL-10. In conclusion, our data suggest that AdipoRon counteracts the cytotoxic effects induced by MS-CSF on SH-SY5Y and U-87 cell lines and that one of the potential molecular underlying mechanisms might occur via reduction in oxidative stress and inflammation. Further in vivo and in vitro studies are essential to confirm whether adiponectin could be a neuro-protectant candidate against neuronal cell injury.
Collapse
Affiliation(s)
- Marta Mallardo
- CEINGE Biotecnologie Avanzate Franco Salvatore, 80145 Naples, Italy
- Dipartimento di Scienze e Tecnologie Ambientali, Biologiche, Farmaceutiche, Università della Campania "Luigi Vanvitelli", 81100 Caserta, Italy
| | - Elisabetta Signoriello
- Centro di Sclerosi Multipla, II Clinica Neurologica, Università della Campania "Luigi Vanvitelli", Via S. Pansini 5, 80131 Naples, Italy
| | - Giacomo Lus
- Centro di Sclerosi Multipla, II Clinica Neurologica, Università della Campania "Luigi Vanvitelli", Via S. Pansini 5, 80131 Naples, Italy
| | - Aurora Daniele
- CEINGE Biotecnologie Avanzate Franco Salvatore, 80145 Naples, Italy
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, "Federico II" Università degli Studi di Napoli, 80131 Naples, Italy
| | - Ersilia Nigro
- CEINGE Biotecnologie Avanzate Franco Salvatore, 80145 Naples, Italy
- Dipartimento di Scienze e Tecnologie Ambientali, Biologiche, Farmaceutiche, Università della Campania "Luigi Vanvitelli", 81100 Caserta, Italy
| |
Collapse
|
3
|
Salwierak-Głośna K, Piątek P, Domowicz M, Świderek-Matysiak M. Effect of Multiple Sclerosis Cerebrospinal Fluid and Oligodendroglia Cell Line Environment on Human Wharton's Jelly Mesenchymal Stem Cells Secretome. Int J Mol Sci 2022; 23:ijms23042177. [PMID: 35216294 PMCID: PMC8878514 DOI: 10.3390/ijms23042177] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 02/11/2022] [Accepted: 02/14/2022] [Indexed: 02/06/2023] Open
Abstract
Multiple sclerosis (MS) is a neurological disorder of autoimmune aetiology. Experimental therapies with the use of mesenchymal stem cells (MSCs) have emerged as a response to the unmet need for new treatment options. The unique immunomodulatory features of stem cells obtained from Wharton’s jelly (WJ-MSCs) make them an interesting research and therapeutic model. Most WJ-MSCs transplants for multiple sclerosis use intrathecal administration. We studied the effect of cerebrospinal fluid (CSF) obtained from MS patients on the secretory activity of WJ-MSCs and broaden this observation with WJ-MSCs interactions with human oligodendroglia cell line (OLs). Analysis of the WJ-MSCs secretory activity with use of Bio-Plex Pro™ Human Cytokine confirmed significant and diverse immunomodulatory potential. Our data reveal rich WJ-MSCs secretome with markedly increased levels of IL-6, IL-8, IP-10 and MCP-1 synthesis and a favourable profile of growth factors. The addition of MS CSF to the WJ-MSCs culture caused depletion of most proteins measured, only IL-12, RANTES and GM-CSF levels were increased. Most cytokines and chemokines decreased their concentrations in WJ-MSCs co-cultured with OLs, only eotaxin and RANTES levels were slightly increased. These results emphasize the spectrum of the immunomodulatory properties of WJ-MSCs and show how those effects can be modulated depending on the transplantation milieu.
Collapse
Affiliation(s)
| | - Paweł Piątek
- Department of Neurology, Medical University of Lodz, 90-419 Lodz, Poland; (K.S.-G.); (P.P.); (M.D.)
- Department of Immunogenetics, Medical University of Lodz, 90-419 Lodz, Poland
| | - Małgorzata Domowicz
- Department of Neurology, Medical University of Lodz, 90-419 Lodz, Poland; (K.S.-G.); (P.P.); (M.D.)
| | - Mariola Świderek-Matysiak
- Department of Neurology, Medical University of Lodz, 90-419 Lodz, Poland; (K.S.-G.); (P.P.); (M.D.)
- Correspondence:
| |
Collapse
|
4
|
Bobinger T, Roeder SS, Spruegel MI, Froehlich K, Beuscher VD, Hoelter P, Lücking H, Corbeil D, Huttner HB. Variation of membrane particle-bound CD133 in cerebrospinal fluid of patients with subarachnoid and intracerebral hemorrhage. J Neurosurg 2021; 134:600-607. [PMID: 31978876 DOI: 10.3171/2019.11.jns191861] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Accepted: 11/25/2019] [Indexed: 11/06/2022]
Abstract
OBJECTIVE Previous studies have demonstrated that human CSF contains membrane particles carrying the stem cell antigenic marker CD133 (prominin-1). Here, the authors analyzed the variation of the amount of these CD133-positive particles in the CSF of patients with subarachnoid hemorrhage (SAH) and intracerebral hemorrhage (ICH). METHODS Consecutive CSF samples from 47 patients with SAH or ICH were compared to 14 healthy control patients. After differential ultracentrifugation of CSF, the membrane particle fraction was separated on gel electrophoresis and its CD133 content was probed by immunoblotting using the mouse monoclonal antibody 80B258 directed against human CD133. The antigen-antibody complexes were detected by chemiluminescence reagents and quantified using human Caco-2 cell extract as positive control with a standardized curve. RESULTS As compared to healthy controls (6.3 ± 0.5 ng of bound CD133 antibody; n = 14), the amount of membrane particle-associated CD133 immunoreactivities was significantly elevated in patients with SAH and ICH (38.2 ± 6.6 ng and 61.3 ± 11.0 ng [p < 0.001] for SAH [n = 18] and ICH [n = 29], respectively). In both groups the CD133 level dropped during the first 7 days (i.e., day 5-7: SAH group, 24.6 ± 10.1 ng [p = 0.06]; ICH group, 25.0 ± 4.8 ng [p = 0.002]). Whereas changes in the amount of CD133-positive membrane particles between admission and day 5-7 were not associated with clinical outcomes in patients with ICH (modified Rankin Scale [mRS] scores 0-3, -30.9 ± 12.8 ng vs mRS scores 4-6, -21.8 ± 10.7 ng; p = 0.239), persistent elevation of CD133 in patients with SAH was related to impaired functional outcome 3 months after ictus (mRS scores 0-2, -29.9 ± 8.1 ng vs mRS scores 3-6, 7.6 ± 20.3 ng; p = 0.027). These data are expressed as the mean ± standard error of the mean (SEM). CONCLUSIONS Levels of membrane particle-associated CD133 in the CSF of patients with SAH and ICH are significantly increased in comparison to healthy patients, and they decline during the hospital stay. Specifically, the persistent elevation of CD133-positive membrane particles within the first week may represent a possible surrogate measure for impaired functional outcome in patients with SAH.
Collapse
Affiliation(s)
| | | | | | | | | | - Philip Hoelter
- 2Neuroradiology, Friedrich-Alexander University Erlangen (FAU); and
| | - Hannes Lücking
- 2Neuroradiology, Friedrich-Alexander University Erlangen (FAU); and
| | - Denis Corbeil
- 3Biotechnology Center (BIOTEC), Technische Universität Dresden, Germany
| | | |
Collapse
|
5
|
Metformin reduces neuronal damage and promotes neuroblast proliferation and differentiation in a cerebral ischemia/reperfusion rat model. Neuroreport 2019; 30:232-240. [PMID: 30614910 DOI: 10.1097/wnr.0000000000001190] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
According to the previous research, metformin, a medication utilized for type 2 diabetes management, inhibits neural aging and reduces infarct size by enhancing angiogenesis in a mouse stroke model. What is more, metformin administration also promotes neural precursor cells proliferation, migration, as well as differentiation for newborn mice with hypoxia-ischemia brain injury. However, whether metformin regulates neurogenesis in an adult rat ischemia/reperfusion (I/R) model remains unclear. The current research found that metformin administration reduced neuronal damage in the CA1 area of hippocampus in a rat model of I/R. The number of neuronal nuclei positive neuron was significantly increased and glial fibrillary acidic protein positive astrocyte became obviously declined in the CA1 region in I/R rats treated with metformin. It was further demonstrated that metformin promoted neuroblasts proliferation and neuronal differentiation in the subgranular zone of the dentate gyrus and inhibited the formation of astrocyte. Our study indicates that activation of endogenous neuroblasts using metformin will become a favorable target in therapeutic intervention of cerebral ischemia injury models.
Collapse
|
6
|
Mazzini L, Gelati M, Profico DC, Sorarù G, Ferrari D, Copetti M, Muzi G, Ricciolini C, Carletti S, Giorgi C, Spera C, Frondizi D, Masiero S, Stecco A, Cisari C, Bersano E, De Marchi F, Sarnelli MF, Querin G, Cantello R, Petruzzelli F, Maglione A, Zalfa C, Binda E, Visioli A, Trombetta D, Torres B, Bernardini L, Gaiani A, Massara M, Paolucci S, Boulis NM, Vescovi AL. Results from Phase I Clinical Trial with Intraspinal Injection of Neural Stem Cells in Amyotrophic Lateral Sclerosis: A Long-Term Outcome. Stem Cells Transl Med 2019; 8:887-897. [PMID: 31104357 PMCID: PMC6708070 DOI: 10.1002/sctm.18-0154] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2018] [Accepted: 02/19/2019] [Indexed: 12/13/2022] Open
Abstract
The main objective of this phase I trial was to assess the feasibility and safety of microtransplanting human neural stem cell (hNSC) lines into the spinal cord of patients with amyotrophic lateral sclerosis (ALS). Eighteen patients with a definite diagnosis of ALS received microinjections of hNSCs into the gray matter tracts of the lumbar or cervical spinal cord. Patients were monitored before and after transplantation by clinical, psychological, neuroradiological, and neurophysiological assessment. For up to 60 months after surgery, none of the patients manifested severe adverse effects or increased disease progression because of the treatment. Eleven patients died, and two underwent tracheotomy as a result of the natural history of the disease. We detected a transitory decrease in progression of ALS Functional Rating Scale Revised, starting within the first month after surgery and up to 4 months after transplantation. Our results show that transplantation of hNSC is a safe procedure that causes no major deleterious effects over the short or long term. This study is the first example of medical transplantation of a highly standardized cell drug product, which can be reproducibly and stably expanded ex vivo, comprising hNSC that are not immortalized, and are derived from the forebrain of the same two donors throughout this entire study as well as across future trials. Our experimental design provides benefits in terms of enhancing both intra‐ and interstudy reproducibility and homogeneity. Given the potential therapeutic effects of the hNSCs, our observations support undertaking future phase II clinical studies in which increased cell dosages are studied in larger cohorts of patients. stem cells translational medicine2019;8:887&897
Collapse
Affiliation(s)
- Letizia Mazzini
- Eastern Piedmont University, "Maggiore della Carità" Hospital, Dipartimento di Neurologia, Novara
| | - Maurizio Gelati
- Laboratorio Cellule Staminali, Cell Factory e Biobanca, Terni Hospital, Italy.,Fondazione IRCCS Casa Sollievo della Sofferenza, Advanced Therapies Production Unit, San Giovanni Rotondo, Foggia, Italy
| | - Daniela Celeste Profico
- Fondazione IRCCS Casa Sollievo della Sofferenza, Advanced Therapies Production Unit, San Giovanni Rotondo, Foggia, Italy
| | - Gianni Sorarù
- Department of Neuroscience, University of Padua, Padua, Italy
| | - Daniela Ferrari
- Biotechnology and Bioscience Department Bicocca University, Milan, Italy
| | - Massimiliano Copetti
- Fondazione IRCCS Casa Sollievo della Sofferenza, Biostatistic Unit, San Giovanni Rotondo, Foggia, Italy
| | - Gianmarco Muzi
- Laboratorio Cellule Staminali, Cell Factory e Biobanca, Terni Hospital, Italy
| | - Claudia Ricciolini
- Laboratorio Cellule Staminali, Cell Factory e Biobanca, Terni Hospital, Italy
| | - Sandro Carletti
- Department of Neurosurgery and Neuroscience, "Santa Maria" Hospital, Terni, Italy
| | - Cesare Giorgi
- Department of Neurosurgery and Neuroscience, "Santa Maria" Hospital, Terni, Italy
| | - Cristina Spera
- Department of Neurosurgery and Neuroscience, "Santa Maria" Hospital, Terni, Italy
| | - Domenico Frondizi
- Department of Neurosurgery and Neuroscience, "Santa Maria" Hospital, Terni, Italy
| | - Stefano Masiero
- Department of Neuroscience, University of Padua, Padua, Italy
| | - Alessandro Stecco
- Department of Diagnostic and Interventional Radiology, "Eastern Piedmont" University, "Maggiore della Carità" Hospital, Novara
| | - Carlo Cisari
- Department of Physical Therapy, "Eastern Piedmont" University, "Maggiore della Carità" Hospital, Novara
| | - Enrica Bersano
- Eastern Piedmont University, "Maggiore della Carità" Hospital, Dipartimento di Neurologia, Novara
| | - Fabiola De Marchi
- Eastern Piedmont University, "Maggiore della Carità" Hospital, Dipartimento di Neurologia, Novara
| | - Maria Francesca Sarnelli
- Eastern Piedmont University, "Maggiore della Carità" Hospital, Dipartimento di Neurologia, Novara
| | - Giorgia Querin
- Department of Neuroscience, University of Padua, Padua, Italy
| | - Roberto Cantello
- Eastern Piedmont University, "Maggiore della Carità" Hospital, Dipartimento di Neurologia, Novara
| | - Francesco Petruzzelli
- Fondazione IRCCS Casa Sollievo della Sofferenza, Obstetrics and Gynaecology Department, San Giovanni Rotondo, Foggia, Italy
| | - Annamaria Maglione
- Fondazione IRCCS Casa Sollievo della Sofferenza, Obstetrics and Gynaecology Department, San Giovanni Rotondo, Foggia, Italy
| | - Cristina Zalfa
- Biotechnology and Bioscience Department Bicocca University, Milan, Italy
| | - Elena Binda
- Fondazione IRCCS Casa Sollievo della Sofferenza, Cancer Stem Cells Unit, San Giovanni Rotondo, Foggia, Italy
| | | | - Domenico Trombetta
- Fondazione IRCCS Casa Sollievo della Sofferenza, Department of Oncology, San Giovanni Rotondo, Foggia, Italy
| | - Barbara Torres
- Fondazione IRCCS Casa Sollievo della Sofferenza, Cytogenetics Unit, San Giovanni Rotondo, Foggia, Italy
| | - Laura Bernardini
- Fondazione IRCCS Casa Sollievo della Sofferenza, Cytogenetics Unit, San Giovanni Rotondo, Foggia, Italy
| | | | - Maurilio Massara
- Eastern Piedmont University, "Maggiore della Carità" Hospital, Dipartimento di Neurologia, Novara
| | - Silvia Paolucci
- Eastern Piedmont University, "Maggiore della Carità" Hospital, Dipartimento di Neurologia, Novara
| | | | - Angelo L Vescovi
- Laboratorio Cellule Staminali, Cell Factory e Biobanca, Terni Hospital, Italy.,Fondazione IRCCS Casa Sollievo della Sofferenza, Advanced Therapies Production Unit, San Giovanni Rotondo, Foggia, Italy.,Biotechnology and Bioscience Department Bicocca University, Milan, Italy
| | | |
Collapse
|
7
|
Stem Cells as Potential Targets of Polyphenols in Multiple Sclerosis and Alzheimer's Disease. BIOMED RESEARCH INTERNATIONAL 2018; 2018:1483791. [PMID: 30112360 PMCID: PMC6077677 DOI: 10.1155/2018/1483791] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/09/2018] [Accepted: 06/19/2018] [Indexed: 12/16/2022]
Abstract
Alzheimer's disease (AD) and multiple sclerosis are major neurodegenerative diseases, which are characterized by the accumulation of abnormal pathogenic proteins due to oxidative stress, mitochondrial dysfunction, impaired autophagy, and pathogens, leading to neurodegeneration and behavioral deficits. Herein, we reviewed the utility of plant polyphenols in regulating proliferation and differentiation of stem cells for inducing brain self-repair in AD and multiple sclerosis. Firstly, we discussed the genetic, physiological, and environmental factors involved in the pathophysiology of both the disorders. Next, we reviewed various stem cell therapies available and how they have proved useful in animal models of AD and multiple sclerosis. Lastly, we discussed how polyphenols utilize the potential of stem cells, either complementing their therapeutic effects or stimulating endogenous and exogenous neurogenesis, against these diseases. We suggest that polyphenols could be a potential candidate for stem cell therapy against neurodegenerative disorders.
Collapse
|
8
|
Ren C, Yin P, Ren N, Wang Z, Wang J, Zhang C, Ge W, Geng D, Wang X. Cerebrospinal fluid-stem cell interactions may pave the path for cell-based therapy in neurological diseases. Stem Cell Res Ther 2018. [PMID: 29523182 PMCID: PMC5845187 DOI: 10.1186/s13287-018-0807-3] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Recent studies have suggested that the regulation of endogenous neural stem cells (NSCs) or transplanting of exogenous nerve cells are the newest and most promising methods for the treatment of dementia and other neurological diseases. The special location and limited number of endogenous NSCs, however, restrict their clinical application. The success in directional differentiation of exogenous stem cells from other tissue sources into neural cells has provided a novel source for NSCs. Study on the relative mechanisms is still at the preliminary stage. Currently the induction methods include: 1) cell growth factor induction; 2) chemical induction; 3) combined growth factor-chemical induction; or 4) other induction methods such as traumatic brain tissue homogenate, gene transfection, traditional Chinese medicine, and coculture induction. Cerebrospinal fluid (CSF), as a natural medium under physiological conditions, contains a variety of progrowth peptide factors that can promote the proliferation and differentiation of mesenchymal stromal cells (MSCs) into neural cells through the corresponding receptors on the cell surface. This suggests that CSF can not only nourish the nerve cells, but also become an effective and suitable inducer to increase the yield of NSCs. However, some other studies believed that CSF contained certain inhibitory components against the differentiation of primary stem cells into mature neural cells. Based on the above background, here we review the relative literature on the influence of the CSF on stem cells in order to provide a more comprehensive reference for the wide clinical application of NSCs in the future.
Collapse
Affiliation(s)
- Chao Ren
- Department of Neurology, Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, 264000, China
| | - Peiyuan Yin
- Department of Blood Supply, Yantai Center Blood Station, Yantai, 264000, China
| | - Neng Ren
- Department of Inervention Therapy, Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221004, China
| | - Zhe Wang
- Department of Clinical Laboratory, Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, 264000, China
| | - Jiahui Wang
- Department of Central Laboratory, Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, 264000, China
| | - Caiyi Zhang
- Department of Neurology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221004, China
| | - Wei Ge
- Department of Neurology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221004, China.
| | - Deqin Geng
- Department of Neurology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221004, China.
| | - Xiaotong Wang
- Department of Neurology, Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, 264000, China.
| |
Collapse
|
9
|
Ji X, Liu H, An C, Wang Y, Zhao H, Zhang Q, Li M, Qi F, Chen Z, Wang X, Wang L. You-Gui pills promote nerve regeneration by regulating netrin1, DCC and Rho family GTPases RhoA, Racl, Cdc42 in C57BL/6 mice with experimental autoimmune encephalomyelitis. JOURNAL OF ETHNOPHARMACOLOGY 2016; 187:123-133. [PMID: 27106785 DOI: 10.1016/j.jep.2016.04.025] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/30/2015] [Revised: 03/14/2016] [Accepted: 04/19/2016] [Indexed: 06/05/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE You-Gui pills (YGPs) are an effective traditional Chinese formula being used clinically for the treatment of multiple sclerosis (MS). Previous studies demonstrated that YGPs exerted the potent neuroprotective effects in murine models of experimental autoimmune encephalomyelitis (EAE), which is an equivalent animal model for multiple sclerosis (MS). However, the mechanism of YGPs functions remained unclear. AIM OF THIS STUDY The aim of this study was to evaluate the therapeutic effect of YGPs in MOG35-55-induced EAE mice and to further elucidate the underlying molecular mechanism. METHODS Female C57BL/6 mice were divided into six groups, including the non-treated EAE model, prednisone acetate- and 1.2, 2.4 or 4.8g/kg YGPs-treated EAE groups, and a normal control group. The EAE model was established by injecting the mice subcutaneously with MOG35-55 antigen. The body weights were measured and the neurological functions were scored in each group. The pathology and morphology of the brain and spinal cord was examined. The expression of MAP-2 was detected by immunofluorescent staining. The levels of netrin1, DCC, RhoA, Rac1, and Cdc42 were assayed by immunohistochemistry, qRT-PCR and Western blot on day 40 post-immunization (PI). RESULTS YGPs treatments significantly reduced neurological function scores in EAE mice, where the inflammatory infiltration was reduced and the axon and myelin damage in both brain and spinal cord was alleviated. In the brain and spinal cord tissues, YGPs increased the expression of neuronal factors MAP-2, netrin1 and DCC. The expression of Rac1 and Cdc42 were increased, while RhoA was reduced following YGPs treatments. CONCLUSION Our results demonstrated that YGPs exhibited a neuroprotective effect on promoting nerve regeneration at the brain and spinal cord in EAE mice induced by MOG35-55. Netrin1, DCC and the Rho family GTPases of RhoA, Racl, Cdc42 were involved in mediating the effects of YGPs on nerve regeneration.
Collapse
MESH Headings
- Animals
- Brain/drug effects
- Brain/pathology
- Brain/ultrastructure
- DCC Receptor
- Drugs, Chinese Herbal/pharmacology
- Drugs, Chinese Herbal/therapeutic use
- Encephalomyelitis, Autoimmune, Experimental/drug therapy
- Encephalomyelitis, Autoimmune, Experimental/metabolism
- Encephalomyelitis, Autoimmune, Experimental/pathology
- Female
- Mice, Inbred C57BL
- Microscopy, Electron, Transmission
- Myelin-Oligodendrocyte Glycoprotein
- Nerve Growth Factors/genetics
- Nerve Growth Factors/metabolism
- Nerve Regeneration/drug effects
- Netrin-1
- Neuroprotective Agents/pharmacology
- Neuroprotective Agents/therapeutic use
- Peptide Fragments
- Phytotherapy
- RNA, Messenger/metabolism
- Receptors, Cell Surface/genetics
- Receptors, Cell Surface/metabolism
- Spinal Cord/drug effects
- Spinal Cord/pathology
- Spinal Cord/ultrastructure
- Tablets
- Tumor Suppressor Proteins/genetics
- Tumor Suppressor Proteins/metabolism
- rho GTP-Binding Proteins/genetics
- rho GTP-Binding Proteins/metabolism
Collapse
Affiliation(s)
- Xiaomin Ji
- School of Traditional Chinese Medicine, Beijing Key Lab of TCM Collateral Disease Theory Research, Capital Medical University, Beijing 100069, PR China
| | - Haolong Liu
- School of Traditional Chinese Medicine, Beijing Key Lab of TCM Collateral Disease Theory Research, Capital Medical University, Beijing 100069, PR China
| | - Chen An
- School of Traditional Chinese Medicine, Beijing Key Lab of TCM Collateral Disease Theory Research, Capital Medical University, Beijing 100069, PR China
| | - Yongqiang Wang
- School of Traditional Chinese Medicine, Beijing Key Lab of TCM Collateral Disease Theory Research, Capital Medical University, Beijing 100069, PR China
| | - Hui Zhao
- School of Traditional Chinese Medicine, Beijing Key Lab of TCM Collateral Disease Theory Research, Capital Medical University, Beijing 100069, PR China
| | - Qiuxia Zhang
- School of Traditional Chinese Medicine, Beijing Key Lab of TCM Collateral Disease Theory Research, Capital Medical University, Beijing 100069, PR China
| | - Ming Li
- School of Traditional Chinese Medicine, Beijing Key Lab of TCM Collateral Disease Theory Research, Capital Medical University, Beijing 100069, PR China
| | - Fang Qi
- School of Traditional Chinese Medicine, Beijing Key Lab of TCM Collateral Disease Theory Research, Capital Medical University, Beijing 100069, PR China
| | - Zhenzhen Chen
- School of Traditional Chinese Medicine, Beijing Key Lab of TCM Collateral Disease Theory Research, Capital Medical University, Beijing 100069, PR China
| | - Xiujuan Wang
- School of Traditional Chinese Medicine, Beijing Key Lab of TCM Collateral Disease Theory Research, Capital Medical University, Beijing 100069, PR China.
| | - Lei Wang
- School of Traditional Chinese Medicine, Beijing Key Lab of TCM Collateral Disease Theory Research, Capital Medical University, Beijing 100069, PR China.
| |
Collapse
|
10
|
Itoh K, Maki T, Lok J, Arai K. Mechanisms of cell-cell interaction in oligodendrogenesis and remyelination after stroke. Brain Res 2015; 1623:135-49. [PMID: 25960351 PMCID: PMC4569526 DOI: 10.1016/j.brainres.2015.04.039] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2015] [Revised: 04/19/2015] [Accepted: 04/20/2015] [Indexed: 12/20/2022]
Abstract
White matter damage is a clinically important aspect of several central nervous system diseases, including stroke. Cerebral white matter primarily consists of axonal bundles ensheathed with myelin secreted by mature oligodendrocytes, which play an important role in neurotransmission between different areas of gray matter. During the acute phase of stroke, damage to oligodendrocytes leads to white matter dysfunction through the loss of myelin. On the contrary, during the chronic phase, white matter components promote an environment, which is favorable for neural repair, vascular remodeling, and remyelination. For effective remyelination to take place, oligodendrocyte precursor cells (OPCs) play critical roles by proliferating and differentiating into mature oligodendrocytes, which help to decrease the burden of axonal injury. Notably, other types of cells contribute to these OPC responses under the ischemic conditions. This mini-review summarizes the non-cell autonomous mechanisms in oligodendrogenesis and remyelination after white matter damage, focusing on how OPCs receive support from their neighboring cells. This article is part of a Special Issue entitled SI: Cell Interactions In Stroke.
Collapse
Affiliation(s)
- Kanako Itoh
- Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| | - Takakuni Maki
- Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| | - Josephine Lok
- Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA; Department of Pediatrics, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| | - Ken Arai
- Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA.
| |
Collapse
|
11
|
Durvasula S, Imitola J. The cerebrospinal fluid-stem cell interactions as target for regenerative therapy in neurological diseases. Stem Cells Dev 2015; 24:145-6. [PMID: 25517016 DOI: 10.1089/scd.2014.0572] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Affiliation(s)
- Sriram Durvasula
- 1 Laboratory for Neural Stem Cells, The Ohio State University Wexner Medical Center , Columbus, Ohio
| | | |
Collapse
|
12
|
Zhu M, Feng Y, Dangelmajer S, Guerrero-Cázares H, Chaichana KL, Smith CL, Levchenko A, Lei T, Quiñones-Hinojosa A. Human cerebrospinal fluid regulates proliferation and migration of stem cells through insulin-like growth factor-1. Stem Cells Dev 2015; 24:160-71. [PMID: 25265906 DOI: 10.1089/scd.2014.0076] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Mesenchymal stem cells (MSCs) and neural progenitor cells (NPCs) have been regarded for their clinical therapeutic potential for central nervous system (CNS) pathologies. Their potential utility is a result of their intrinsic ability to repair damaged tissues, deliver therapeutic proteins, and migrate to sites of pathology within the brain. However, it remains unclear whether the CNS promotes any changes in these potential therapeutic cells, which would be critical to understand before clinical application. A major component of the CNS is cerebrospinal fluid (CSF). Therefore, the aim of this study was to evaluate the influence that human CSF has on the function of human adipose-derived MSCs (hAMSCs) and human fetal-derived NPCs (hfNPCs) in regard to cell proliferation, survival, and migration. This study demonstrated that human noncancerous CSF promoted proliferation and inhibited apoptosis of hAMSCs and hfNPCs. Preculturing these stem cells in human CSF also increased their migratory speed and distance traveled. Furthermore, insulin-like growth factor-1 (IGF-1) in human CSF enhanced the migration capacity and increased the expression of C-X-C chemokine receptor type 4 (CXCR4) in both stem cell types. These current findings highlight a simple and natural way in which human CSF can enhance the proliferation, migration, and viability of human exogenous primary hAMSCs and hfNPCs. This study may provide insight into improving the clinical efficacy of stem cells for the treatment of CNS pathologies.
Collapse
Affiliation(s)
- Mingxin Zhu
- 1 Department of Neurosurgery and Oncology, Johns Hopkins University School of Medicine , Baltimore, Maryland
| | | | | | | | | | | | | | | | | |
Collapse
|
13
|
Marro BS, Blanc CA, Loring JF, Cahalan MD, Lane TE. Promoting remyelination: utilizing a viral model of demyelination to assess cell-based therapies. Expert Rev Neurother 2015; 14:1169-79. [PMID: 25245576 DOI: 10.1586/14737175.2014.955854] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Multiple sclerosis (MS) is a chronic inflammatory disease of the CNS. While a broad range of therapeutics effectively reduce the incidence of focal white matter inflammation and plaque formation for patients with relapse-remitting forms of MS, a challenge within the field is to develop therapies that allow for axonal protection and remyelination. In the last decade, growing interest has focused on utilizing neural precursor cells (NPCs) to promote remyelination. To understand how NPCs function in chronic demyelinating environments, several excellent pre-clinical mouse models have been developed. One well accepted model is infection of susceptible mice with neurotropic variants of mouse hepatitis virus (MHV) that undergo chronic demyelination exhibiting clinical and histopathologic similarities to MS patients. Combined with the possibility that an environmental agent such as a virus could trigger MS, the MHV model of demyelination presents a relevant mouse model to assess the therapeutic potential of NPCs transplanted into an environment in which inflammatory-mediated demyelination is established.
Collapse
Affiliation(s)
- Brett S Marro
- Department of Molecular Biology and Biochemistry, University of California, Irvine 92697, USA
| | | | | | | | | |
Collapse
|
14
|
Haines JD, Vidaurre OG, Zhang F, Riffo-Campos ÁL, Castillo J, Casanova B, Casaccia P, Lopez-Rodas G. Multiple sclerosis patient-derived CSF induces transcriptional changes in proliferating oligodendrocyte progenitors. Mult Scler 2015; 21:1655-69. [PMID: 25948622 DOI: 10.1177/1352458515573094] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2014] [Accepted: 01/25/2015] [Indexed: 12/13/2022]
Abstract
BACKGROUND Cerebrospinal fluid (CSF) is in contact with brain parenchyma and ventricles, and its composition might influence the cellular physiology of oligodendrocyte progenitor cells (OPCs) thereby contributing to multiple sclerosis (MS) disease pathogenesis. OBJECTIVE To identify the transcriptional changes that distinguish the transcriptional response induced in proliferating rat OPCs upon exposure to CSF from primary progressive multiple sclerosis (PPMS) or relapsing remitting multiple sclerosis (RRMS) patients and other neurological controls. METHODS We performed gene microarray analysis of OPCs exposed to CSF from neurological controls, or definitive RRMS or PPMS disease course. Results were confirmed by quantitative reverse transcriptase polymerase chain reaction, immunocytochemistry and western blot of cultured cells, and validated in human brain specimens. RESULTS We identified common and unique oligodendrocyte genes for each treatment group. Exposure to CSF from PPMS uniquely induced branching of cultured progenitors and related transcriptional changes, including upregulation (P<0.05) of the adhesion molecule GALECTIN-3/Lgals3, which was also detected at the protein level in brain specimens from PPMS patients. This pattern of gene expression was distinct from the transcriptional programme of oligodendrocyte differentiation during development. CONCLUSIONS Despite evidence of morphological differentiation induced by exposure to CSF of PPMS patients, the overall transcriptional response elicited in cultured OPCs was consistent with the activation of an aberrant transcriptional programme.
Collapse
Affiliation(s)
- Jeffery D Haines
- Department of Neuroscience, Genetics and Genomics, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Oscar G Vidaurre
- Department of Neuroscience, Genetics and Genomics, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Fan Zhang
- Department of Neuroscience, Genetics and Genomics, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Ángela L Riffo-Campos
- Department of Biochemistry and Molecular Biology, University of Valencia, and Institute of Health Research INCLIVA, Valencia, Spain
| | - Josefa Castillo
- Department of Biochemistry and Molecular Biology, University of Valencia, and Institute of Health Research INCLIVA, Valencia, Spain
| | | | - Patrizia Casaccia
- Department of Neuroscience, Genetics and Genomics, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Gerardo Lopez-Rodas
- Department of Biochemistry and Molecular Biology, University of Valencia, 46100 Burjassot, Valencia, Spain
| |
Collapse
|
15
|
Xu J, Nash RJ, Frey TK. Cellular responses to Sindbis virus infection of neural progenitors derived from human embryonic stem cells. BMC Res Notes 2014; 7:757. [PMID: 25343994 PMCID: PMC4307679 DOI: 10.1186/1756-0500-7-757] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2014] [Accepted: 10/10/2014] [Indexed: 11/12/2022] Open
Abstract
Background Sindbis virus (SINV) causes age-dependent encephalitis in mice, and therefore serves as a model to study viral encephalitis. SINV is used as a vector for the delivery of genes into selected neural stem cell lines; however, the toxicity and side effects of this vector have rarely been discussed. In this context, we investigated the cellular responses of human embryonic stem cell (hESCs) derived neural progenitors (hNPCs) to SINV infection by assessing susceptibility of the cells to SINV infection, analyzing the effect of infection on cell proliferation and cell death, and examining the impact of SINV infection on hNPCs markers of stemness. Findings We found that hNPCs are highly susceptible to SINV infection. Upon infection, the viruses induced apoptosis to hNPCs while not affecting the expression of cell proliferation markers. Lastly, SINV infections result in significant changes in the expression of key regulators of hNPCs’ plasticity and homeostasis. Conclusion The robust and versatile signaling, proliferation, and other cell responses of hESCs-derived hNPCs to virus infection demonstrated that it is a good model to study the pathogenesis of viral-induced neurodevelopmental and degenerative diseases. On the other hand, the toxicity of SINV to hNPCs cells cannot be ignored, and therefore extra care should be taken when using SINV as a vector to deliver genes into human stem cell lines.
Collapse
Affiliation(s)
| | | | - Teryl K Frey
- Department of Biology, Georgia State University, Atlanta, GA, USA.
| |
Collapse
|
16
|
Cristofanilli M, Rosenthal H, Cymring B, Gratch D, Pagano B, Xie B, Sadiq SA. Progressive multiple sclerosis cerebrospinal fluid induces inflammatory demyelination, axonal loss, and astrogliosis in mice. Exp Neurol 2014; 261:620-32. [PMID: 25111532 DOI: 10.1016/j.expneurol.2014.07.020] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2014] [Revised: 07/24/2014] [Accepted: 07/29/2014] [Indexed: 01/10/2023]
Abstract
Multiple sclerosis (MS) is an autoimmune disease characterized by inflammatory demyelination and neurodegeneration throughout the CNS, which lead over time to a condition of irreversible functional decline known as progressive MS. Currently, there are no satisfactory treatments for this condition because the mechanisms that underlie disease progression are not well understood. This is partly due to the lack of a specific animal model that represents progressive MS. We investigated the effects of intracerebroventricular injections of cerebrospinal fluid (CSF) derived from untreated primary progressive (PPMS), secondary progressive (SPMS), and relapsing/remitting (RRMS) MS patients into mice. We found discrete inflammatory demyelinating lesions containing macrophages, B cell and T cell infiltrates in the brains of animals injected with CSF from patients with progressive MS. These lesions were rarely found in animals injected with RRMS-CSF and never in those treated with control-CSF. Animals that developed brain lesions also presented extensive inflammation in their spinal cord. However, discrete spinal cord lesions were rare and only seen in animals injected with PPMS-CSF. Axonal loss and astrogliosis were seen within the lesions following the initial demyelination. In addition, Th17 cell activity was enhanced in the CNS and in lymph nodes of progressive MS-CSF injected animals compared to controls. Furthermore, CSF derived from MS patients who were clinically stable following therapy had greatly diminished capacity to induce CNS lesions in mice. Finally, we provided evidence suggesting that differential expression of pro-inflammatory cytokines present in the progressive MS CSF might be involved in the observed mouse pathology. Our data suggests that the agent(s) responsible for the demyelination and neurodegeneration characteristic of progressive MS is present in patient CSF and is amenable to further characterization in experimental models of the disease.
Collapse
Affiliation(s)
| | | | - Barbara Cymring
- Tisch MS Research Center of New York, New York, NY 10019, USA
| | - Daniel Gratch
- Tisch MS Research Center of New York, New York, NY 10019, USA
| | - Benjamin Pagano
- Tisch MS Research Center of New York, New York, NY 10019, USA
| | - Boxun Xie
- Tisch MS Research Center of New York, New York, NY 10019, USA
| | - Saud A Sadiq
- Tisch MS Research Center of New York, New York, NY 10019, USA.
| |
Collapse
|