1
|
Zhuo Y, Wang F, Lv Q, Fang C. Dissolving microneedles: Drug delivery and disease treatment. Colloids Surf B Biointerfaces 2025; 250:114571. [PMID: 39983455 DOI: 10.1016/j.colsurfb.2025.114571] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2024] [Revised: 02/10/2025] [Accepted: 02/15/2025] [Indexed: 02/23/2025]
Abstract
Traditional transdermal drug delivery methods are often plagued by technical inefficiencies, limited absorption, and the potential for adverse reactions. In contrast, dissolving microneedles (DMNs) offer a novel approach to transdermal drug delivery by effectively merging the benefits of subcutaneous injection with those of conventional transdermal methods. These microneedles dissolve completely within the body, releasing the encapsulated antigen without leaving any sharp remnants. Furthermore, DMNs overcome the limitations of traditional transdermal patches, which are restricted to delivering only small molecule drugs. By facilitating the efficient transdermal absorption of large molecules, DMNs enable precise and painless disease treatment. With advantages such as effective delivery, safety, controllable administration, DMNs hold significant promise in the fields of disease treatment and drug delivery. This article explores the substrate materials, preparation techniques, characterization methods, and current applications of DMNs. We also discuss the current challenges and obstacles faced by DMNs. Finally, we outline potential future research directions for DMNs, aiming to provide a theoretical reference for researchers involved in their preparation and application.
Collapse
Affiliation(s)
- Yanling Zhuo
- College of Veterinary Medicine, Hunan Agricultural University, Changsha 410128, China; College of Intelligent Agriculture, Yulin Normal University, Yulin 537000, China
| | - Fangyue Wang
- Chengdu University of Traditional Chinese Medicine, Chengdu 610072, China
| | - Qizhuang Lv
- College of Intelligent Agriculture, Yulin Normal University, Yulin 537000, China; Guangxi Key Laboratory of Agricultural Resources Chemistry and Biotechnology, Yulin 537000, China.
| | - Chunyan Fang
- Institute of Quality Standard and Testing Technology for Agro-Products, Sichuan Academy of Agricultural Sciences, Chengdu 610066, China.
| |
Collapse
|
2
|
Kumar U. Cannabinoids: Role in Neurological Diseases and Psychiatric Disorders. Int J Mol Sci 2024; 26:152. [PMID: 39796008 PMCID: PMC11720483 DOI: 10.3390/ijms26010152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Revised: 12/20/2024] [Accepted: 12/22/2024] [Indexed: 01/13/2025] Open
Abstract
An impact of legalization and decriminalization of marijuana is the gradual increase in the use of cannabis for recreational purposes, which poses a potential threat to society and healthcare systems worldwide. However, the discovery of receptor subtypes, endogenous endocannabinoids, and enzymes involved in synthesis and degradation, as well as pharmacological characterization of receptors, has led to exploration of the use of cannabis in multiple peripheral and central pathological conditions. The role of cannabis in the modulation of crucial events involving perturbed physiological functions and disease progression, including apoptosis, inflammation, oxidative stress, perturbed mitochondrial function, and the impaired immune system, indicates medicinal values. These events are involved in most neurological diseases and prompt the gradual progression of the disease. At present, several synthetic agonists and antagonists, in addition to more than 70 phytocannabinoids, are available with distinct efficacy as a therapeutic alternative in different pathological conditions. The present review aims to describe the use of cannabis in neurological diseases and psychiatric disorders.
Collapse
Affiliation(s)
- Ujendra Kumar
- Faculty of Pharmaceutical Sciences, The University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| |
Collapse
|
3
|
Lan XY, Liang XS, Cao MX, Qin HM, Chu CY, Boltze J, Li S. NCAM mimetic peptide P2 synergizes with bone marrow mesenchymal stem cells in promoting functional recovery after stroke. J Cereb Blood Flow Metab 2024; 44:1128-1144. [PMID: 38230663 PMCID: PMC11179606 DOI: 10.1177/0271678x241226482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 11/07/2023] [Accepted: 12/08/2023] [Indexed: 01/18/2024]
Abstract
The neural cell adhesion molecule (NCAM) promotes neural development and regeneration. Whether NCAM mimetic peptides could synergize with bone marrow mesenchymal stem cells (BMSCs) in stroke treatment deserves investigation. We found that the NCAM mimetic peptide P2 promoted BMSC proliferation, migration, and neurotrophic factor expression, protected neurons from oxygen-glucose deprivation through ERK and PI3K/AKT activation and anti-apoptotic mechanisms in vitro. Following middle cerebral artery occlusion (MCAO) in rats, P2 alone or in combination with BMSCs inhibited neuronal apoptosis and induced the phosphorylation of ERK and AKT. P2 combined with BMSCs enhanced neurotrophic factor expression and BMSC proliferation in the ischemic boundary zone. Moreover, combined P2 and BMSC therapy induced translocation of nuclear factor erythroid 2-related factor, upregulated heme oxygenase-1 expression, reduced infarct volume, and increased functional recovery as compared to monotreatments. Treatment with LY294002 (PI3K inhibitor) and PD98059 (ERK inhibitor) decreased the neuroprotective effects of combined P2 and BMSC therapy in MCAO rats. Collectively, P2 is neuroprotective while P2 and BMSCs work synergistically to improve functional outcomes after ischemic stroke, which may be attributed to mechanisms involving enhanced BMSC proliferation and neurotrophic factor release, anti-apoptosis, and PI3K/AKT and ERK pathways activation.
Collapse
Affiliation(s)
- Xiao-Yan Lan
- Department of Neurology, Dalian Municipal Central Hospital, Dalian, China
| | - Xue-Song Liang
- Department of Neurology and Psychiatry, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Ming-Xuan Cao
- Department of Neurology and Psychiatry, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Hua-Min Qin
- Department of Pathology, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Cheng-Yan Chu
- Department of Neurology, Dalian Municipal Central Hospital, Dalian, China
| | - Johannes Boltze
- School of Life Sciences, University of Warwick, Coventry, UK
| | - Shen Li
- Department of Neurology and Psychiatry, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
- Beijing Institute of Brain Disorders, Capital Medical University, Beijing, China
| |
Collapse
|
4
|
Moniche F, Cabezas-Rodriguez JA, Valverde R, Escudero-Martinez I, Lebrato-Hernandez L, Pardo-Galiana B, Ainz L, Medina-Rodriguez M, de la Torre J, Escamilla-Gomez V, Ortega-Quintanilla J, Zapata-Arriaza E, de Albóniga-Chindurza A, Mancha F, Gamero MA, Perez S, Espinosa-Rosso R, Forero-Diaz L, Moya M, Piñero P, Calderón-Cabrera C, Nogueras S, Jimenez R, Martin V, Delgado F, Ochoa-Sepúlveda JJ, Quijano B, Mata R, Santos-González M, Carmona-Sanchez G, Herrera C, Gonzalez A, Montaner J. Safety and efficacy of intra-arterial bone marrow mononuclear cell transplantation in patients with acute ischaemic stroke in Spain (IBIS trial): a phase 2, randomised, open-label, standard-of-care controlled, multicentre trial. Lancet Neurol 2023; 22:137-146. [PMID: 36681446 DOI: 10.1016/s1474-4422(22)00526-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 11/14/2022] [Accepted: 12/01/2022] [Indexed: 01/21/2023]
Abstract
BACKGROUND Pilot clinical trials have shown the safety of intra-arterial bone marrow mononuclear cells (BMMNCs) in stroke. However, the efficacy of different doses of intra-arterial BMMNCs in patients with acute stroke has not been tested in a randomised clinical trial. We aimed to show safety and efficacy of two different doses of autologous intra-arterial BMMNC transplantation in patients with acute stroke. METHODS The IBIS trial was a multicentre phase 2, randomised, controlled, investigator-initiated, assessor-blinded, clinical trial, in four stroke centres in Spain. We included patients (aged 18-80 years) with a non-lacunar, middle cerebral artery ischaemic stroke within 1-7 days from stroke onset and with a National Institutes of Health Stroke Scale score of 6-20. We randomly assigned patients (2:1:1) with a computer-generated randomisation sequence to standard of care (control group) or intra-arterial injection of autologous BMMNCs at one of two different doses (2 × 106 BMMNCs/kg or 5 × 106 BMMNCs/kg). The primary efficacy outcome was the proportion of patients with modified Rankin Scale scores of 0-2 at 180 days in the intention-to-treat population, comparing each BMMNC dose group and the pooled BMMNC group versus the control group. The primary safety endpoint was the proportion of serious adverse events. This trial was registered at ClinicalTrials.gov, NCT02178657 and is completed. FINDINGS Between April 1, 2015, and May 20, 2021, we assessed 114 patients for eligibility. We randomly assigned 77 (68%) patients: 38 (49%) to the control group, 20 (26%) to the low-dose BMMNC group, and 19 (25%) the high-dose BMMNC group. The mean age of participants was 62·4 years (SD 12·7), 46 (60%) were men, 31 (40%) were women, all were White, and 63 (82%) received thrombectomy. The median NIHSS score before randomisation was 12 (IQR 9-15), with intra-arterial BMMNC injection done a median of 6 days (4-7) after stroke onset. The primary efficacy outcome occurred in 14 (39%) patients in the control group versus ten (50%) in the low-dose group (adjusted odds ratio 2·08 [95% CI 0·55-7·85]; p=0·28), eight (44%) in the high-dose group (1·89 [0·52-6·96]; p=0·33), and 18 (47%) in the pooled BMMNC group (2·22 [0·72-6·85]; p=0·16). We found no differences in the proportion of patients who had adverse events or dose-related events, but two patients had a groin haematoma after cell injection in the low-dose BMMNC group. INTERPRETATION Intra-arterial BMMNCs were safe in patients with acute ischaemic stroke, but we found no significant improvement at 180 days on the mRS. Further clinical trials are warranted to investigate whether improvements might be possible at different timepoints. FUNDING Instituto de Salud Carlos III co-funded by the European Regional Development Fund/European Social Fund, Mutua Madrileña, and the Regional Ministry of Health of Andalusia.
Collapse
Affiliation(s)
- Francisco Moniche
- Department of Neurology, Virgen del Rocío University Hospital, Seville, Spain; Neurovascular Lab, Instituto de Biomedicina de Sevilla-IBiS, Seville, Spain.
| | | | - Roberto Valverde
- Department of Neurology, Department of Radiology, Reina Sofía University Hospital, Cordoba, Spain
| | - Irene Escudero-Martinez
- Department of Neurology, Virgen del Rocío University Hospital, Seville, Spain; Neurovascular Lab, Instituto de Biomedicina de Sevilla-IBiS, Seville, Spain
| | | | | | - Leire Ainz
- Department of Neurology, Virgen del Rocío University Hospital, Seville, Spain
| | - Manuel Medina-Rodriguez
- Department of Neurology, Virgen del Rocío University Hospital, Seville, Spain; Neurovascular Lab, Instituto de Biomedicina de Sevilla-IBiS, Seville, Spain
| | - Javier de la Torre
- Department of Neurology, Virgen del Rocío University Hospital, Seville, Spain
| | | | | | - Elena Zapata-Arriaza
- Interventional Neuroradiology, Virgen del Rocío University Hospital, Seville, Spain
| | | | - Fernando Mancha
- Neurovascular Lab, Instituto de Biomedicina de Sevilla-IBiS, Seville, Spain
| | - Miguel-Angel Gamero
- Department of Neurology, Virgen Macarena University Hospital, Seville, Spain
| | - Soledad Perez
- Department of Neurology, Virgen Macarena University Hospital, Seville, Spain
| | | | - Lucia Forero-Diaz
- Department of Neurology, Puerta del Mar University Hospital, Cadiz, Spain
| | - Miguel Moya
- Department of Neurology, Puerta del Mar University Hospital, Cadiz, Spain
| | - Pilar Piñero
- Department of Radiology, Virgen del Rocío University Hospital, Seville, Spain
| | | | - Sonia Nogueras
- Cell Therapy Unit, Reina Sofía University Hospital, IMIBIC, University of Córdoba, Córdoba, Spain
| | - Rosario Jimenez
- Cell Therapy Unit, Reina Sofía University Hospital, IMIBIC, University of Córdoba, Córdoba, Spain
| | - Vanesa Martin
- Department of Hematology, Reina Sofía University Hospital, IMIBIC, University of Córdoba, Córdoba, Spain; Cell Therapy Unit, Reina Sofía University Hospital, IMIBIC, University of Córdoba, Córdoba, Spain
| | - Fernando Delgado
- Interventional Neuroradiology, Department of Radiology, Reina Sofía University Hospital, Cordoba, Spain
| | | | - Blanca Quijano
- Coordination Unit of the Andalusian Network for the design and translation of Advanced Therapies, Seville, Spain
| | - Rosario Mata
- Coordination Unit of the Andalusian Network for the design and translation of Advanced Therapies, Seville, Spain
| | - Monica Santos-González
- Production and Reprogramming Cell Unit of Seville, Andalusian Network for the Design and Translation of Advanced Therapies, Seville, Spain; Centro de Transfusiones, Tejidos y Células de Sevilla (CTTS), Fundación Pública Andaluza para la Gestión de la Investigación en Salud en Sevilla (FISEVI), Seville, Spain
| | - Gloria Carmona-Sanchez
- Coordination Unit of the Andalusian Network for the design and translation of Advanced Therapies, Seville, Spain; Production and Reprogramming Cell Unit of Seville, Andalusian Network for the Design and Translation of Advanced Therapies, Seville, Spain
| | - Concha Herrera
- Department of Hematology, Reina Sofía University Hospital, IMIBIC, University of Córdoba, Córdoba, Spain; Cell Therapy Unit, Reina Sofía University Hospital, IMIBIC, University of Córdoba, Córdoba, Spain
| | - Alejandro Gonzalez
- Interventional Neuroradiology, Virgen del Rocío University Hospital, Seville, Spain
| | - Joan Montaner
- Neurovascular Lab, Instituto de Biomedicina de Sevilla-IBiS, Seville, Spain; Department of Neurology, Virgen Macarena University Hospital, Seville, Spain
| |
Collapse
|
5
|
Ghozy S, Reda A, Varney J, Elhawary AS, Shah J, Murry K, Sobeeh MG, Nayak SS, Azzam AY, Brinjikji W, Kadirvel R, Kallmes DF. Neuroprotection in Acute Ischemic Stroke: A Battle Against the Biology of Nature. Front Neurol 2022; 13:870141. [PMID: 35711268 PMCID: PMC9195142 DOI: 10.3389/fneur.2022.870141] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2022] [Accepted: 04/21/2022] [Indexed: 12/22/2022] Open
Abstract
Stroke is the second most common cause of global death following coronary artery disease. Time is crucial in managing stroke to reduce the rapidly progressing insult of the ischemic penumbra and the serious neurologic deficits that might follow it. Strokes are mainly either hemorrhagic or ischemic, with ischemic being the most common of all types of strokes. Thrombolytic therapy with recombinant tissue plasminogen activator and endovascular thrombectomy are the main types of management of acute ischemic stroke (AIS). In addition, there is a vital need for neuroprotection in the setting of AIS. Neuroprotective agents are important to investigate as they may reduce mortality, lessen disability, and improve quality of life after AIS. In our review, we will discuss the main types of management and the different modalities of neuroprotection, their mechanisms of action, and evidence of their effectiveness after ischemic stroke.
Collapse
Affiliation(s)
- Sherief Ghozy
- Department of Neuroradiology, Mayo Clinic, Rochester, MN, United States.,Nuffield Department of Primary Care Health Sciences and Department for Continuing Education (EBHC Program), Oxford University, Oxford, United Kingdom
| | - Abdullah Reda
- Faculty of Medicine, Al-Azhar University, Cairo, Egypt
| | - Joseph Varney
- School of Medicine, American University of the Caribbean, Philipsburg, Sint Maarten
| | | | - Jaffer Shah
- Medical Research Center, Kateb University, Kabul, Afghanistan
| | | | - Mohamed Gomaa Sobeeh
- Faculty of Physical Therapy, Sinai University, Cairo, Egypt.,Faculty of Physical Therapy, Cairo University, Giza, Egypt
| | - Sandeep S Nayak
- Department of Internal Medicine, NYC Health + Hospitals/Metropolitan, New York, NY, United States
| | - Ahmed Y Azzam
- Faculty of Medicine, October 6 University, Giza, Egypt
| | - Waleed Brinjikji
- Department of Neurosurgery, Mayo Clinic Rochester, Rochester, MN, United States
| | | | - David F Kallmes
- Department of Neuroradiology, Mayo Clinic, Rochester, MN, United States
| |
Collapse
|
6
|
Rizzo SA, Bartley O, Rosser AE, Newland B. Oxygen-glucose deprivation in neurons: implications for cell transplantation therapies. Prog Neurobiol 2021; 205:102126. [PMID: 34339808 DOI: 10.1016/j.pneurobio.2021.102126] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Revised: 07/16/2021] [Accepted: 07/29/2021] [Indexed: 12/25/2022]
Abstract
Cell replacement therapies hold the potential to restore neuronal networks compromised by neurodegenerative diseases (such as Parkinson's disease or Huntington's disease), or focal tissue damage (via a stroke or spinal cord injury). Despite some promising results achieved to date, transplanted cells typically exhibit poor survival in the central nervous system, thus limiting therapeutic efficacy of the graft. Although cell death post-transplantation is likely to be multifactorial in causality, growing evidence suggests that the lack of vascularisation at the graft site, and the resulting ischemic host environment, may play a fundamental role in the fate of grafted cells. Herein, we summarise data showing how the deprivation of either oxygen, glucose, or both in combination, impacts the survival of neurons and review strategies which may improve graft survival in the central nervous system.
Collapse
Affiliation(s)
| | - Oliver Bartley
- Brain Repair Group, School of Biosciences, Cardiff University, Cardiff, CF10 3AX, Wales, UK
| | - Anne E Rosser
- Brain Repair Group, School of Biosciences, Cardiff University, Cardiff, CF10 3AX, Wales, UK; Neuroscience and Mental Health Institute and B.R.A.I.N Unit, Cardiff University, School of Medicine, Hadyn Ellis Building, Maindy Road, CF24 4HQ, Cardiff, UK
| | - Ben Newland
- School of Pharmacy and Pharmaceutical Sciences, Cardiff University, CF10 3NB, Wales, UK; Leibniz Institute for Polymer Research Dresden (IPF), Hohe Straße 6, 01069, Dresden, Germany.
| |
Collapse
|
7
|
Zhou G, Wang Y, Gao S, Fu X, Cao Y, Peng Y, Zhuang J, Hu J, Shao A, Wang L. Potential Mechanisms and Perspectives in Ischemic Stroke Treatment Using Stem Cell Therapies. Front Cell Dev Biol 2021; 9:646927. [PMID: 33869200 PMCID: PMC8047216 DOI: 10.3389/fcell.2021.646927] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Accepted: 03/05/2021] [Indexed: 12/12/2022] Open
Abstract
Ischemic stroke (IS) remains one of the major causes of death and disability due to the limited ability of central nervous system cells to regenerate and differentiate. Although several advances have been made in stroke therapies in the last decades, there are only a few approaches available to improve IS outcome. In the acute phase of IS, mechanical thrombectomy and the administration of tissue plasminogen activator have been widely used, while aspirin or clopidogrel represents the main therapy used in the subacute or chronic phase. However, in most cases, stroke patients fail to achieve satisfactory functional recovery under the treatments mentioned above. Recently, cell therapy, especially stem cell therapy, has been considered as a novel and potential therapeutic strategy to improve stroke outcome through mechanisms, including cell differentiation, cell replacement, immunomodulation, neural circuit reconstruction, and protective factor release. Different stem cell types, such as mesenchymal stem cells, marrow mononuclear cells, and neural stem cells, have also been considered for stroke therapy. In recent years, many clinical and preclinical studies on cell therapy have been carried out, and numerous results have shown that cell therapy has bright prospects in the treatment of stroke. However, some cell therapy issues are not yet fully understood, such as its optimal parameters including cell type choice, cell doses, and injection routes; therefore, a closer relationship between basic and clinical research is needed. In this review, the role of cell therapy in stroke treatment and its mechanisms was summarized, as well as the function of different stem cell types in stroke treatment and the clinical trials using stem cell therapy to cure stroke, to reveal future insights on stroke-related cell therapy, and to guide further studies.
Collapse
Affiliation(s)
- Guoyang Zhou
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Yongjie Wang
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Shiqi Gao
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Xiongjie Fu
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Yang Cao
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Yucong Peng
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Jianfeng Zhuang
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Junwen Hu
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Anwen Shao
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Lin Wang
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
8
|
Lee S, Kim OJ, Lee KO, Jung H, Oh SH, Kim NK. Enhancing the Therapeutic Potential of CCL2-Overexpressing Mesenchymal Stem Cells in Acute Stroke. Int J Mol Sci 2020; 21:ijms21207795. [PMID: 33096826 PMCID: PMC7588958 DOI: 10.3390/ijms21207795] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 10/19/2020] [Accepted: 10/19/2020] [Indexed: 01/15/2023] Open
Abstract
Although intravenous administration of mesenchymal stem cells (MSCs) is effective for experimental stroke, low engraftment and the limited functional capacity of transplanted cells are critical hurdles for clinical applications. C-C motif chemokine ligand 2 (CCL2) is associated with neurological repair after stroke and delivery of various cells into the brain via CCL2/CCR2 (CCL2 receptor) interaction. In this study, after CCL2-overexpressing human umbilical cord-derived MSCs (hUC-MSCs) were intravenously transplanted with mannitol in rats with middle cerebral arterial occlusion, we compared the differences between four different treatment groups: mannitol + CCL2-overexpressing hUC-MSCs (CCL2-MSC), mannitol + naïve hUC-MSCs (M-MSC), mannitol only, and control. At four-weeks post-transplantation, the CCL2-MSC group showed significantly better functional recovery and smaller stroke volume relative to the other groups. Additionally, we observed upregulated levels of CCR2 in acute ischemic brain and the increase of migrated stem cells into these areas in the CCL2-MSC group relative to the M-MSC. Moreover, the CCL2-MSC group displayed increased angiogenesis and endogenous neurogenesis, decreased neuro-inflammation but with increased healing-process inflammatory cells relative to other groups. These findings indicated that CCL2-overexpressing hUC-MSCs showed better functional recovery relative to naïve hUC-MSCs according to the increased migration of these cells into brain areas of higher CCR2 expression, thereby promoting subsequent endogenous brain repair.
Collapse
Affiliation(s)
- Sanghun Lee
- Department of Neurology, CHA Bundang Medical Center, CHA University, Seongnam 13496, Korea; (S.L.); (K.O.L.); (H.J.); (S.-H.O.)
| | - Ok Joon Kim
- Department of Neurology, CHA Bundang Medical Center, CHA University, Seongnam 13496, Korea; (S.L.); (K.O.L.); (H.J.); (S.-H.O.)
- Correspondence: ; Tel.: +82-31-780-5481; Fax: +82-31-780-5269
| | - Kee Ook Lee
- Department of Neurology, CHA Bundang Medical Center, CHA University, Seongnam 13496, Korea; (S.L.); (K.O.L.); (H.J.); (S.-H.O.)
| | - Hyeju Jung
- Department of Neurology, CHA Bundang Medical Center, CHA University, Seongnam 13496, Korea; (S.L.); (K.O.L.); (H.J.); (S.-H.O.)
| | - Seung-Hun Oh
- Department of Neurology, CHA Bundang Medical Center, CHA University, Seongnam 13496, Korea; (S.L.); (K.O.L.); (H.J.); (S.-H.O.)
| | - Nam Keun Kim
- Institute for Clinical Research, CHA Bundang Medical Center, CHA University, Seongnam 13488, Korea;
| |
Collapse
|
9
|
Wang K, Rong L, Wei X, Zhang Q, Xiao L. The effectiveness of various cytotherapeutic strategies for the treatment of ischemic stroke: a Bayesian network meta-analysis of randomized controlled trials. Neurol Sci 2020; 41:1705-1717. [PMID: 32130557 DOI: 10.1007/s10072-020-04312-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Accepted: 02/20/2020] [Indexed: 12/13/2022]
Abstract
BACKGROUND Cytotherapy is a potential treatment for ischemic stroke (IS) patients but lacks uniform procedures. We aimed to assess the impact of the time of intervention, cell type, dose, and route of administration on the clinical effects by network meta-analysis. METHODS We searched public electronic databases through July 7, 2019. Bayesian network meta-analyses were performed to compare differences among different cytotherapeutic strategies. RESULTS Cytotherapy can significantly improve patients' activity of daily living according to the modified Rankin Scale (standard mean difference (SMD) - 0.81; 95% confidence interval (CI) - 1.58, - 0.03; p = 0.0417) and Barthel Index (SMD 0.67; 95% CI 0.05, 1.30; p = 0.036) results as well as improve neurological recovery (SMD - 0.93; 95% CI - 1.29, - 0.57; p < 0.001). Network meta-analysis showed that the intra-arterial injection of large amounts of mononuclear cells (NCs) or aldehyde dehydrogenase (ALDH)-positive cells was beneficial for improving patients' activity of daily living, while CD34+ cells through intracerebral injection had an advantage in the recovery of injured nerve function. Intravenous injection of mesenchymal stem cells (MSCs) or endothelial progenitor cells (EPCs) was beneficial in reducing mortality and serious adverse event (SAE) onset. CONCLUSIONS In the subacute stage, the intra-arterial injection of NCs or ALDH cells improves patients' activity of daily living. Additionally, CD34+ cells through intracerebral injection had an advantage in the recovery of injured nerve function even in the chronic stage. Intravenous injection of MSCs or EPCs is a safety delivery route that can reduce mortality and SAE onset. However, further clinical studies are still needed to confirm these results.
Collapse
Affiliation(s)
- Kai Wang
- Department of Neurology, Second Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, 221006, China
| | - Liangqun Rong
- Department of Neurology, Second Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, 221006, China.
- , No. 32, Meijian Road, Quanshan District, Xuzhou, Jiangsu, China.
| | - Xiue Wei
- Department of Neurology, Second Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, 221006, China
| | - Qingxiu Zhang
- Department of Neurology, Second Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, 221006, China
| | - Lijie Xiao
- Department of Neurology, Second Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, 221006, China
| |
Collapse
|
10
|
Bonsack B, Corey S, Shear A, Heyck M, Cozene B, Sadanandan N, Zhang H, Gonzales-Portillo B, Sheyner M, Borlongan CV. Mesenchymal stem cell therapy alleviates the neuroinflammation associated with acquired brain injury. CNS Neurosci Ther 2020; 26:603-615. [PMID: 32356605 PMCID: PMC7248547 DOI: 10.1111/cns.13378] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Revised: 03/28/2020] [Accepted: 03/29/2020] [Indexed: 01/01/2023] Open
Abstract
Ischemic stroke and traumatic brain injury (TBI) comprise two particularly prevalent and costly examples of acquired brain injury (ABI). Following stroke or TBI, primary cell death and secondary cell death closely model disease progression and worsen outcomes. Mounting evidence indicates that long‐term neuroinflammation extensively exacerbates the secondary deterioration of brain structure and function. Due to their immunomodulatory and regenerative properties, mesenchymal stem cell transplants have emerged as a promising approach to treating this facet of stroke and TBI pathology. In this review, we summarize the classification of cell death in ABI and discuss the prominent role of inflammation. We then consider the efficacy of bone marrow–derived mesenchymal stem/stromal cell (BM‐MSC) transplantation as a therapy for these injuries. Finally, we examine recent laboratory and clinical studies utilizing transplanted BM‐MSCs as antiinflammatory and neurorestorative treatments for stroke and TBI. Clinical trials of BM‐MSC transplants for stroke and TBI support their promising protective and regenerative properties. Future research is needed to allow for better comparison among trials and to elaborate on the emerging area of cell‐based combination treatments.
Collapse
Affiliation(s)
- Brooke Bonsack
- Department of Neurosurgery and Brain Repair, University of South Florida, Tampa, FL, USA
| | - Sydney Corey
- Department of Neurosurgery and Brain Repair, University of South Florida, Tampa, FL, USA
| | - Alex Shear
- Department of Neurosurgery and Brain Repair, University of South Florida, Tampa, FL, USA
| | - Matt Heyck
- Department of Neurosurgery and Brain Repair, University of South Florida, Tampa, FL, USA
| | - Blaise Cozene
- Department of Neurosurgery and Brain Repair, University of South Florida, Tampa, FL, USA
| | - Nadia Sadanandan
- Department of Neurosurgery and Brain Repair, University of South Florida, Tampa, FL, USA
| | - Henry Zhang
- Department of Neurosurgery and Brain Repair, University of South Florida, Tampa, FL, USA
| | | | - Michael Sheyner
- Department of Neurosurgery and Brain Repair, University of South Florida, Tampa, FL, USA
| | - Cesar V Borlongan
- Department of Neurosurgery and Brain Repair, University of South Florida, Tampa, FL, USA
| |
Collapse
|
11
|
Krucoff MO, Miller JP, Saxena T, Bellamkonda R, Rahimpour S, Harward SC, Lad SP, Turner DA. Toward Functional Restoration of the Central Nervous System: A Review of Translational Neuroscience Principles. Neurosurgery 2020; 84:30-40. [PMID: 29800461 DOI: 10.1093/neuros/nyy128] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2018] [Accepted: 03/15/2018] [Indexed: 01/09/2023] Open
Abstract
Injury to the central nervous system (CNS) can leave patients with devastating neurological deficits that may permanently impair independence and diminish quality of life. Recent insights into how the CNS responds to injury and reacts to critically timed interventions are being translated into clinical applications that have the capacity to drastically improve outcomes for patients suffering from permanent neurological deficits due to spinal cord injury, stroke, or other CNS disorders. The translation of such knowledge into practical and impactful treatments involves the strategic collaboration between neurosurgeons, clinicians, therapists, scientists, and industry. Therefore, a common understanding of key neuroscientific principles is crucial. Conceptually, current approaches to CNS revitalization can be divided by scale into macroscopic (systems-circuitry) and microscopic (cellular-molecular). Here we review both emerging and well-established tenets that are being utilized to enhance CNS recovery on both levels, and we explore the role of neurosurgeons in developing therapies moving forward. Key principles include plasticity-driven functional recovery, cellular signaling mechanisms in axonal sprouting, critical timing for recovery after injury, and mechanisms of action underlying cellular replacement strategies. We then discuss integrative approaches aimed at synergizing interventions across scales, and we make recommendations for the basis of future clinical trial design. Ultimately, we argue that strategic modulation of microscopic cellular behavior within a macroscopic framework of functional circuitry re-establishment should provide the foundation for most neural restoration strategies, and the early involvement of neurosurgeons in the process will be crucial to successful clinical translation.
Collapse
Affiliation(s)
- Max O Krucoff
- Department of Neurosurgery, Duke University Medical Center, Durham, North Carolina
| | - Jonathan P Miller
- Department of Neurosurgery, Case Western Reserve University, Cleve-land, Ohio
| | - Tarun Saxena
- Department of Biomedical Engineering, Duke University, Durham, North Carolina
| | - Ravi Bellamkonda
- Department of Biomedical Engineering, Duke University, Durham, North Carolina
| | - Shervin Rahimpour
- Department of Neurosurgery, Duke University Medical Center, Durham, North Carolina
| | - Stephen C Harward
- Department of Neurosurgery, Duke University Medical Center, Durham, North Carolina
| | - Shivanand P Lad
- Department of Neurosurgery, Duke University Medical Center, Durham, North Carolina.,Department of Mechan-ical Engineering and Material Sciences, Pratt School of Engineering, Duke Uni-versity, Durham, North Carolina.,Duke Institute for Brain Sciences, Duke Univer-sity, Durham, North Carolina.,Research and Surgery Services, Durham Veterans Affairs Medical Center, Durham, North Carolina
| | - Dennis A Turner
- Department of Neurosurgery, Duke University Medical Center, Durham, North Carolina.,Department of Biomedical Engineering, Duke University, Durham, North Carolina.,Depart-ment of Neurobiology, Duke University, Durham, North Carolina.,Research and Surgery Services, Durham Veterans Affairs Medical Center, Durham, North Carolina
| |
Collapse
|
12
|
Zuo L, Feng Q, Han Y, Chen M, Guo M, Liu Z, Cheng Y, Li G. Therapeutic effect on experimental acute cerebral infarction is enhanced after nanoceria labeling of human umbilical cord mesenchymal stem cells. Ther Adv Neurol Disord 2019; 12:1756286419859725. [PMID: 31431809 PMCID: PMC6685115 DOI: 10.1177/1756286419859725] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2019] [Accepted: 06/03/2019] [Indexed: 12/15/2022] Open
Abstract
Background: Therapeutic applications of stem cells, especially mesenchymal stem cells, were once regarded as a promising therapy for mitigating acute cerebral infarction. Unfortunately, all the stem cell clinical trials have been futile. A new stroke therapeutic strategy of combining stem cells with nanotechnology has recently gained significant attention. The objective of this study was to evaluate the application of cerium oxide nanoparticle (nanoceria)-labeled human umbilical cord mesenchymal stem cells (HucMSCs) for stroke therapy. Methods: In our study, cerium oxide nanoparticles were precovered with hyaluronic acid before labeling HucMSCs and the synergistic effects from both HucMSCs and cerium oxide nanoparticles were analyzed in in vivo and in vitro experiments Results: The nanoceria-labeled HucMSCs combined advantages from both sides, including the capacity for inflammatory modulation of HucMSCs and the antioxidant effects of nanoceria. Compared with either HucMSCs or nanoceria individually, nanoceria-labeled HucMSCs exerted significantly enhanced capacities after gaining combined antioxidant and anti-inflammatory effects. Conclusion: Our findings suggest a novel strategy with effective and well-tolerated applications of stem cells for acute cerebral infarction therapy after modification of cells with nanomaterials.
Collapse
Affiliation(s)
- Lian Zuo
- Department of Neurology, East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Qishuai Feng
- Department of Neurology, East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yingying Han
- Department of Neurology, East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Mengwei Chen
- East Hospital, The Institute for Biomedical Engineering and Nano Science, Tongji University School of Medicine, Shanghai, China
| | - Mengruo Guo
- Department of Neurology, East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Zhongmin Liu
- Department of Intensive Care Center, East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
| | - Yu Cheng
- East Hospital; The Institute for Biomedical Engineering & Nano Science, Tongji University School of Medicine, Shanghai, 200120, China
| | - Gang Li
- Department of Neurology,East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
| |
Collapse
|
13
|
Zhang Y, Deng H, Hu Y, Pan C, Wu G, Li Q, Tang Z. Adipose-derived mesenchymal stem cells stereotactic transplantation alleviate brain edema from intracerebral hemorrhage. J Cell Biochem 2019; 120:14372-14382. [PMID: 30963640 DOI: 10.1002/jcb.28693] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Revised: 02/25/2019] [Accepted: 02/28/2019] [Indexed: 02/06/2023]
Abstract
Adipose-derived mesenchymal stromal cells (ADSCs) exhibited high potential in tissue repair and regeneration, and it has been proved that ADSCs could protect brain cells from apoptosis and maintaining blood-brain barrier stability after cerebral vascular disease. In this study, we evaluated the therapeutic potential and mechanism of ADSCs stereotactic transplantation in intracerebral hemorrhage (ICH) mice model and hemin-treated astrocytes. Mice were divided into three groups: sham group, ICH + PBS group, and ICH + ADSC group. Mice in ICH + ADSC group received ADSCs cell suspension stereotactic transplantation into the area beside the bleeding region. Astrocytes were divided into three groups: control group, hemin group, and hemin + ADSC group. Astrocytes in hemin + ADSC group were cultured in ADSCs-astrocyte no-contact coculture system and treated with 30 μM hemin solution. The results showed that ADSCs stereotactic transplantation improved functional outcomes and reduced cell apoptosis after ICH. Moreover, ADSCs stereotactic transplantation could alleviate brain edema and inflammation and AQP4 protein expression contributed to the alleviation of brain edema. In addition, mitogen-activated protein kinase (MAPK) pathways, including p38/MAPK pathway and c-Jun N-terminal kinase pathway, were involved in AQP4 modulation by ADSCs transplantation in ICH. In conclusion, ADSCs transplantation could alleviate the nervous tissue injury, reduce cell apoptosis, and relieve brain edema in ICH. And the edema regulation effect of ADSCs transplantation is associated with inhibition of inflammation and AQP4 protein expression.
Collapse
Affiliation(s)
- Ye Zhang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Hong Deng
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Yang Hu
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Chao Pan
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Guofeng Wu
- Department of Emergency, Affiliated Hospital of Guizhou Medical University, Guiyang, People's Republic of China
| | - Qi Li
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, People's Republic of China
| | - Zhouping Tang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| |
Collapse
|
14
|
Finch-Edmondson M, Morgan C, Hunt RW, Novak I. Emergent Prophylactic, Reparative and Restorative Brain Interventions for Infants Born Preterm With Cerebral Palsy. Front Physiol 2019; 10:15. [PMID: 30745876 PMCID: PMC6360173 DOI: 10.3389/fphys.2019.00015] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Accepted: 01/08/2019] [Indexed: 12/13/2022] Open
Abstract
Worldwide, an estimated 15 million babies are born preterm (<37 weeks' gestation) every year. Despite significant improvements in survival rates, preterm infants often face a lifetime of neurodevelopmental disability including cognitive, behavioral, and motor impairments. Indeed, prematurity remains the largest risk factor for the development of cerebral palsy. The developing brain of the preterm infant is particularly fragile; preterm babies exhibit varying severities of cerebral palsy arising from reductions in both cerebral white and gray matter volumes, as well as altered brain microstructure and connectivity. Current intensive care therapies aim to optimize cardiovascular and respiratory function to protect the brain from injury by preserving oxygenation and blood flow. If a brain injury does occur, definitive diagnosis of cerebral palsy in the first few hours and weeks of life is difficult, especially when the lesions are subtle and not apparent on cranial ultrasound. However, early diagnosis of mildly affected infants is critical, because these are the patients most likely to respond to emergent treatments inducing neuroplasticity via high-intensity motor training programs and regenerative therapies involving stem cells. A current controversy is whether to test universal treatment in all infants at risk of brain injury, accepting that some patients never required treatment, because the perceived potential benefits outweigh the risk of harm. Versus, waiting for a diagnosis before commencing targeted treatment for infants with a brain injury, and potentially missing the therapeutic window. In this review, we discuss the emerging prophylactic, reparative, and restorative brain interventions for infants born preterm, who are at high risk of developing cerebral palsy. We examine the current evidence, considering the timing of the intervention with relation to the proposed mechanism/s of action. Finally, we consider the development of novel markers of preterm brain injury, which will undoubtedly lead to improved diagnostic and prognostic capability, and more accurate instruments to assess the efficacy of emerging interventions for this most vulnerable group of infants.
Collapse
Affiliation(s)
- Megan Finch-Edmondson
- The Discipline of Child and Adolescent Health, The Children's Hospital at Westmead Clinical School, The University of Sydney Medical School, Sydney, NSW, Australia
- Cerebral Palsy Alliance Research Institute, The University of Sydney, Sydney, NSW, Australia
| | - Catherine Morgan
- The Discipline of Child and Adolescent Health, The Children's Hospital at Westmead Clinical School, The University of Sydney Medical School, Sydney, NSW, Australia
- Cerebral Palsy Alliance Research Institute, The University of Sydney, Sydney, NSW, Australia
| | - Rod W. Hunt
- Department of Neonatal Medicine, The Royal Children's Hospital, Melbourne, VIC, Australia
- Department of Paediatrics, University of Melbourne, Melbourne, VIC, Australia
- Neonatal Research, Murdoch Children's Research Institute, Melbourne, VIC, Australia
- Department of Obstetrics and Gynaecology, Monash University, Melbourne, VIC, Australia
| | - Iona Novak
- The Discipline of Child and Adolescent Health, The Children's Hospital at Westmead Clinical School, The University of Sydney Medical School, Sydney, NSW, Australia
- Cerebral Palsy Alliance Research Institute, The University of Sydney, Sydney, NSW, Australia
| |
Collapse
|
15
|
Narayanan DL, Phadke SR. Concepts, Utility and Limitations of Cord Blood Banking: What Clinicians Need to Know. Indian J Pediatr 2019; 86:44-48. [PMID: 29556970 DOI: 10.1007/s12098-018-2651-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Accepted: 02/27/2018] [Indexed: 11/29/2022]
Abstract
Stem cell transplantation and cord blood banking have received much popularity among general public and medical professionals in the recent past. But information about the scientific aspects, its utility and limitations is incomplete amongst laypersons as well as many medical practitioners. Stem cells differ from all other types of cells in the human body because of their ability to multiply in order to self perpetuate and differentiate into specialized cells. Stems cells could be totipotent, multipotent, pluripotent, oligopotent or unipotent depending on the type of cells that can arise or differentiate from them. Umbilical cord blood serves as a potent source of hematopoeitic stem cells and is being used to treat various disorders like blood cancers, hemoglobinopathies and immunodeficiency disorders for which hematological stem cell transplantation is the standard of care. Cord blood can be collected at ease, without any major complications and has a lower incidence of graft vs. host reaction compared to bone marrow cells or peripheral blood cells. Both public and private banks have been established for collection and storage of umbilical cord blood. However, false claims and misleading commercial advertisements about the use of umbilical cord blood stem cells for the treatment of a variety of conditions ranging from neuromuscular disorders to cosmetic benefits are widespread and create unrealistic expectations in laypersons and clinicians. Many clinicians and laypersons are unaware of the limitations of cord blood banking, as in treating a genetic disorder by autologous cord blood transplant. Knowledge and awareness about the scientific indications of cord blood stem cell transplantation and realistic expectations about the utility of cord blood among medical practitioners are essential for providing accurate information to laypersons before they decide to preserve umbilical cord blood in private banks and thus prevent malpractice.
Collapse
Affiliation(s)
- Dhanya Lakshmi Narayanan
- Department of Medical Genetics, Nizam's Institute of Medical Sciences, Punjagutta, Hyderabad, India
| | - Shubha R Phadke
- Department of Medical Genetics, Sanjay Gandhi Post Graduate Institute of Medical Sciences, Lucknow, India.
| |
Collapse
|
16
|
Van de Putte D, Demarquay C, Van Daele E, Moussa L, Vanhove C, Benderitter M, Ceelen W, Pattyn P, Mathieu N. Adipose-Derived Mesenchymal Stromal Cells Improve the Healing of Colonic Anastomoses Following High Dose of Irradiation Through Anti-Inflammatory and Angiogenic Processes. Cell Transplant 2018; 26:1919-1930. [PMID: 29390877 PMCID: PMC5802630 DOI: 10.1177/0963689717721515] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Cancer patients treated with radiotherapy (RT) could develop severe late side effects that affect their quality of life. Long-term bowel complications after RT are mainly characterized by a transmural fibrosis that could lead to intestinal obstruction. Today, surgical resection is the only effective treatment. However, preoperative RT increases the risk of anastomotic leakage. In this study, we attempted to use mesenchymal stromal cells from adipose tissue (Ad-MSCs) to improve colonic anastomosis after high-dose irradiation. MSCs were isolated from the subcutaneous fat of rats, amplified in vitro, and characterized by flow cytometry. An animal model of late radiation side effects was induced by local irradiation of the colon. Colonic anastomosis was performed 4 wk after irradiation. It was analyzed another 4 wk later (i.e., 8 wk after irradiation). The Ad-MSC-treated group received injections several times before and after the surgical procedure. The therapeutic benefit of the Ad-MSC treatment was determined by colonoscopy and histology. The inflammatory process was investigated using Fluorine-182-Fluoro-2-Deoxy-d-Glucose Positron Emission Tomography and Computed Tomography (18F-FDG-PET/CT) imaging and macrophage infiltrate analyses. Vascular density was assessed using immunohistochemistry. Results show that Ad-MSC treatment reduces ulcer size, increases mucosal vascular density, and limits hemorrhage. We also determined that 1 Ad-MSC injection limits the inflammatory process, as evaluated through 18F-FDG-PET-CT (at 4 wk), with a greater proportion of type 2 macrophages after iterative cell injections (8 wk). In conclusion, Ad-MSC injections promote anastomotic healing in an irradiated colon through enhanced vessel formation and reduced inflammation. This study also determined parameters that could be improved in further investigations.
Collapse
Affiliation(s)
- Dirk Van de Putte
- 1 Department of Pediatric and Gastrointestinal Surgery, Ghent University Hospital, Ghent, Belgium
| | - Christelle Demarquay
- 2 Institut de Radioprotection et de Sûreté Nucléaire (IRSN), Fontenay-aux-Roses, France
| | - Elke Van Daele
- 1 Department of Pediatric and Gastrointestinal Surgery, Ghent University Hospital, Ghent, Belgium
| | - Lara Moussa
- 2 Institut de Radioprotection et de Sûreté Nucléaire (IRSN), Fontenay-aux-Roses, France
| | | | - Marc Benderitter
- 2 Institut de Radioprotection et de Sûreté Nucléaire (IRSN), Fontenay-aux-Roses, France
| | - Wim Ceelen
- 1 Department of Pediatric and Gastrointestinal Surgery, Ghent University Hospital, Ghent, Belgium.,4 Cancer Research Institute Ghent (CRIG), Ghent, Belgium
| | - Piet Pattyn
- 1 Department of Pediatric and Gastrointestinal Surgery, Ghent University Hospital, Ghent, Belgium
| | - Noëlle Mathieu
- 2 Institut de Radioprotection et de Sûreté Nucléaire (IRSN), Fontenay-aux-Roses, France
| |
Collapse
|
17
|
Qian Y, Han Q, Chen W, Song J, Zhao X, Ouyang Y, Yuan W, Fan C. Platelet-Rich Plasma Derived Growth Factors Contribute to Stem Cell Differentiation in Musculoskeletal Regeneration. Front Chem 2017; 5:89. [PMID: 29164105 PMCID: PMC5671651 DOI: 10.3389/fchem.2017.00089] [Citation(s) in RCA: 91] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2017] [Accepted: 10/16/2017] [Indexed: 12/24/2022] Open
Abstract
Stem cell treatment and platelet-rich plasma (PRP) therapy are two significant issues in regenerative medicine. Stem cells such as bone marrow mesenchymal stem cells, adipose-derived stem cells and periodontal ligament stem cells can be successfully applied in the field of tissue regeneration. PRP, a natural product isolated from whole blood, can secrete multiple growth factors (GFs) for regulating physiological activities. These GFs can stimulate proliferation and differentiation of different stem cells in injury models. Therefore, combination of both agents receives wide expectations in regenerative medicine, especially in bone, cartilage and tendon repair. In this review, we thoroughly discussed the interaction and underlying mechanisms of PRP derived GFs with stem cells, and assessed their functions in cell differentiation for musculoskeletal regeneration.
Collapse
Affiliation(s)
- Yun Qian
- Department of Orthopedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China.,Shanghai Sixth People's Hospital East Campus, Shanghai University of Medicine and Health, Shanghai, China
| | - Qixin Han
- Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Wei Chen
- Department of Orthopedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China.,Shanghai Sixth People's Hospital East Campus, Shanghai University of Medicine and Health, Shanghai, China
| | - Jialin Song
- Department of Orthopedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China.,Shanghai Sixth People's Hospital East Campus, Shanghai University of Medicine and Health, Shanghai, China
| | - Xiaotian Zhao
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai, China
| | - Yuanming Ouyang
- Department of Orthopedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China.,Shanghai Sixth People's Hospital East Campus, Shanghai University of Medicine and Health, Shanghai, China
| | - Weien Yuan
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai, China
| | - Cunyi Fan
- Department of Orthopedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| |
Collapse
|
18
|
Sammali E, Alia C, Vegliante G, Colombo V, Giordano N, Pischiutta F, Boncoraglio GB, Barilani M, Lazzari L, Caleo M, De Simoni MG, Gaipa G, Citerio G, Zanier ER. Intravenous infusion of human bone marrow mesenchymal stromal cells promotes functional recovery and neuroplasticity after ischemic stroke in mice. Sci Rep 2017; 7:6962. [PMID: 28761170 PMCID: PMC5537246 DOI: 10.1038/s41598-017-07274-w] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2017] [Accepted: 06/27/2017] [Indexed: 12/13/2022] Open
Abstract
Transplantation of human bone marrow mesenchymal stromal cells (hBM-MSC) promotes functional recovery after stroke in animal models, but the mechanisms underlying these effects remain incompletely understood. We tested the efficacy of Good Manufacturing Practices (GMP) compliant hBM-MSC, injected intravenously 3.5 hours after injury in mice subjected to transient middle cerebral artery occlusion (tMCAo). We addressed whether hBM-MSC are efficacious and if this efficacy is associated with cortical circuit reorganization using neuroanatomical analysis of GABAergic neurons (parvalbumin; PV-positive cells) and perineuronal nets (PNN), a specialized extracellular matrix structure which acts as an inhibitor of neural plasticity. tMCAo mice receiving hBM-MSC, showed early and lasting improvement of sensorimotor and cognitive functions compared to control tMCAo mice. Furthermore, 5 weeks post-tMCAo, hBM-MSC induced a significant rescue of ipsilateral cortical neurons; an increased proportion of PV-positive neurons in the perilesional cortex, suggesting GABAergic interneurons preservation; and a lower percentage of PV-positive cells surrounded by PNN, indicating an enhanced plastic potential of the perilesional cortex. These results show that hBM-MSC improve functional recovery and stimulate neuroprotection after stroke. Moreover, the downregulation of “plasticity brakes” such as PNN suggests that hBM-MSC treatment stimulates plasticity and formation of new connections in the perilesional cortex.
Collapse
Affiliation(s)
- Eliana Sammali
- Department of Neuroscience, IRCCS - Istituto di Ricerche Farmacologiche Mario Negri, Via La Masa,19, 20156, Milano, Italy.,Department of Cerebrovascular Diseases, Fondazione IRCCS - Istituto Neurologico Carlo Besta, Milano, Italy
| | - Claudia Alia
- Neuroscience Institute, CNR, Pisa, Italy.,Scuola Normale Superiore, Pisa, Italy
| | - Gloria Vegliante
- Department of Neuroscience, IRCCS - Istituto di Ricerche Farmacologiche Mario Negri, Via La Masa,19, 20156, Milano, Italy
| | - Valentina Colombo
- Laboratory for Cell and Gene Therapy "Stefano Verri", ASST-Monza, San Gerardo Hospital, Monza, Italy.,Tettamanti Research Center, Pediatric Department, University of Milano-Bicocca, Monza, Italy
| | - Nadia Giordano
- Neuroscience Institute, CNR, Pisa, Italy.,Scuola Normale Superiore, Pisa, Italy
| | - Francesca Pischiutta
- Department of Neuroscience, IRCCS - Istituto di Ricerche Farmacologiche Mario Negri, Via La Masa,19, 20156, Milano, Italy
| | - Giorgio B Boncoraglio
- Department of Cerebrovascular Diseases, Fondazione IRCCS - Istituto Neurologico Carlo Besta, Milano, Italy
| | - Mario Barilani
- Cell Factory, Unit of Cell Therapy and Cryobiology, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Via F. Sforza 35, 20122, Milano, Italy
| | - Lorenza Lazzari
- Cell Factory, Unit of Cell Therapy and Cryobiology, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Via F. Sforza 35, 20122, Milano, Italy
| | | | - Maria-Grazia De Simoni
- Department of Neuroscience, IRCCS - Istituto di Ricerche Farmacologiche Mario Negri, Via La Masa,19, 20156, Milano, Italy
| | - Giuseppe Gaipa
- Laboratory for Cell and Gene Therapy "Stefano Verri", ASST-Monza, San Gerardo Hospital, Monza, Italy.,Tettamanti Research Center, Pediatric Department, University of Milano-Bicocca, Monza, Italy
| | - Giuseppe Citerio
- School of Medicine and Surgery, University of Milano-Bicocca, Milano, Italy.,Neurointensive Care, ASST-Monza, San Gerardo Hospital, Monza, Italy
| | - Elisa R Zanier
- Department of Neuroscience, IRCCS - Istituto di Ricerche Farmacologiche Mario Negri, Via La Masa,19, 20156, Milano, Italy.
| |
Collapse
|
19
|
Stem cell therapy for abrogating stroke-induced neuroinflammation and relevant secondary cell death mechanisms. Prog Neurobiol 2017; 158:94-131. [PMID: 28743464 DOI: 10.1016/j.pneurobio.2017.07.004] [Citation(s) in RCA: 186] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2017] [Revised: 07/18/2017] [Accepted: 07/18/2017] [Indexed: 12/13/2022]
Abstract
Ischemic stroke is a leading cause of death worldwide. A key secondary cell death mechanism mediating neurological damage following the initial episode of ischemic stroke is the upregulation of endogenous neuroinflammatory processes to levels that destroy hypoxic tissue local to the area of insult, induce apoptosis, and initiate a feedback loop of inflammatory cascades that can expand the region of damage. Stem cell therapy has emerged as an experimental treatment for stroke, and accumulating evidence supports the therapeutic efficacy of stem cells to abrogate stroke-induced inflammation. In this review, we investigate clinically relevant stem cell types, such as hematopoietic stem cells (HSCs), mesenchymal stem cells (MSCs), endothelial progenitor cells (EPCs), very small embryonic-like stem cells (VSELs), neural stem cells (NSCs), extraembryonic stem cells, adipose tissue-derived stem cells, breast milk-derived stem cells, menstrual blood-derived stem cells, dental tissue-derived stem cells, induced pluripotent stem cells (iPSCs), teratocarcinoma-derived Ntera2/D1 neuron-like cells (NT2N), c-mycER(TAM) modified NSCs (CTX0E03), and notch-transfected mesenchymal stromal cells (SB623), comparing their potential efficacy to sequester stroke-induced neuroinflammation and their feasibility as translational clinical cell sources. To this end, we highlight that MSCs, with a proven track record of safety and efficacy as a transplantable cell for hematologic diseases, stand as an attractive cell type that confers superior anti-inflammatory effects in stroke both in vitro and in vivo. That stem cells can mount a robust anti-inflammatory action against stroke complements the regenerative processes of cell replacement and neurotrophic factor secretion conventionally ascribed to cell-based therapy in neurological disorders.
Collapse
|
20
|
Garbuzova-Davis S, Haller E, Lin R, Borlongan CV. Intravenously Transplanted Human Bone Marrow Endothelial Progenitor Cells Engraft Within Brain Capillaries, Preserve Mitochondrial Morphology, and Display Pinocytotic Activity Toward Blood-Brain Barrier Repair in Ischemic Stroke Rats. Stem Cells 2017; 35:1246-1258. [PMID: 28142208 DOI: 10.1002/stem.2578] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2016] [Revised: 01/05/2017] [Accepted: 01/12/2017] [Indexed: 01/01/2023]
Abstract
Stroke is a life-threatening disease with limited therapeutic options. Cell therapy has emerged as an experimental stroke treatment. Blood-brain barrier (BBB) impairment is a key pathological manifestation of ischemic stroke, and barrier repair is an innovative target for neurorestoration in stroke. Here, we evaluated via electron microscopy the ability of transplanted human bone marrow endothelial progenitor cells (hBMEPCs) to repair the BBB in adult Sprague-Dawley rats subjected to transient middle cerebral artery occlusion (tMCAO). β-galactosidase prelabeled hBMEPCs were intravenously transplanted 48 hours post-tMCAO. Ultrastructural analysis of microvessels in nontransplant stroke rats revealed typical BBB pathology. At 5 days post-transplantation with hBMEPCs, stroke rats displayed widespread vascular repair in bilateral striatum and motor cortex, characterized by robust cell engraftment within capillaries. hBMEPC transplanted stroke rats exhibited near normal morphology of endothelial cells (ECs), pericytes, and astrocytes, without detectable perivascular edema. Near normal morphology of mitochondria was also detected in ECs and perivascular astrocytes from transplanted stroke rats. Equally notable, we observed numerous pinocytic vesicles within engrafted cells. Robust engraftment and intricate functionality of transplanted hBMEPCs likely abrogated stroke-altered vasculature. Preserving mitochondria and augmenting pinocytosis in cell-based therapeutics represent a new neurorestorative mechanism in BBB repair for stroke. Stem Cells 2017;35:1246-1258.
Collapse
Affiliation(s)
- Svitlana Garbuzova-Davis
- Center of Excellence for Aging & Brain Repair.,Department of Neurosurgery and Brain Repair.,Department of Molecular Pharmacology and Physiology.,Department of Pathology and Cell Biology, Morsani College of Medicine
| | - Edward Haller
- Department of Integrative Biology, University of South Florida, Tampa, Florida, USA
| | - Roger Lin
- Center of Excellence for Aging & Brain Repair
| | - Cesario V Borlongan
- Center of Excellence for Aging & Brain Repair.,Department of Neurosurgery and Brain Repair
| |
Collapse
|
21
|
Detante O, Moisan A, Hommel M, Jaillard A. Controlled clinical trials of cell therapy in stroke: Meta-analysis at six months after treatment. Int J Stroke 2017; 12:748-751. [DOI: 10.1177/1747493017696098] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Background Cell therapy is promising in experimental studies and has been assessed only in a few studies on humans. Aims To evaluate the effect of cell therapy in humans. Methods We included clinical trials with a control group that reported safety and efficacy six months following treatment. Quality was evaluated and clinical scales data were extracted. Quantitative analysis was based on the standardized means difference (SMD). Among 28 trials published from 1995 to 2016, nine studies (194 patients; 191 controls) were eligible. Publication biases were assessed with the funnel plot and pre-specified explanatory variables were tested with a group analysis and a meta-regression. Results The overall quality was moderate. Cell therapy had a positive effect on the outcome (SMD: 0.57, 95% CI: 0.22–0.92; p = 0.002). The sensitivity analysis showed an upper level of effect size of 0.81 (95% CI: 0.34–1.27; p = 0.001) and a lower level of 0.455 (95% CI: 0.04–0.87; p = 0.03). None of the pre-specified explanatory variable was significantly correlated to outcome: age, ratio infarction/hemorrhage, delay from stroke to treatment, route of administration, cell type, randomization, and blinded outcome assessment. The significant heterogeneity (p = 0.03) was not explained by publication biases (p = 0.09) and was more likely due to methodological and quality differences between the trials. Conclusions This result suggests that cell therapy is beneficial in stroke and is expected to help in the designing of stem cells controlled clinical trials (CCT) in large populations.
Collapse
Affiliation(s)
- Olivier Detante
- Stroke Unit, Department of Neurology, University Hospital of Grenoble, Grenoble, France
- Inserm, U 836, Grenoble, France
- Grenoble Institute of Neurosciences, Grenoble-Alpes University, Grenoble, France
| | - Anaïck Moisan
- Unité de Thérapie et d'Ingénierie Cellulaire – EFS Rhône-Alpes-Auvergne, Saint Ismier, France
- Institute for Advanced Biosciences UGA, Grenoble, France
| | - Marc Hommel
- Department of Research, University Hospital of Grenoble, Grenoble, France
- Grenoble-Alpes University, AGEIS EA 7407
| | - Assia Jaillard
- Department of Research, University Hospital of Grenoble, Grenoble, France
- Grenoble-Alpes University, AGEIS EA 7407
- 3T-MRI Research Unit, IRMAGE, University Hospital of Grenoble, Grenoble, France
| |
Collapse
|
22
|
Feng J, Chen X, Shen J. Reactive nitrogen species as therapeutic targets for autophagy: implication for ischemic stroke. Expert Opin Ther Targets 2017; 21:305-317. [DOI: 10.1080/14728222.2017.1281250] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
- Jinghan Feng
- School of Chinese Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
- The University of Hong Kong-Shenzhen Institute of Research and Innovation (HKU-SIRI), Shenzhen, China
| | - Xingmiao Chen
- School of Chinese Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
- The University of Hong Kong-Shenzhen Institute of Research and Innovation (HKU-SIRI), Shenzhen, China
| | - Jiangang Shen
- School of Chinese Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
- The University of Hong Kong-Shenzhen Institute of Research and Innovation (HKU-SIRI), Shenzhen, China
| |
Collapse
|
23
|
Unsworth DJ, Mathias JL, Dorstyn DS. Cell therapies administered in the chronic phase after stroke: a meta-analysis examining safety and efficacy. Regen Med 2017; 12:91-108. [DOI: 10.2217/rme-2016-0082] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Aim: To assess the safety and efficacy of cell therapies for chronic stroke. Methodology: Five databases were searched for treatments administered >90 days post-stroke. Reporting quality, adherence to research guidelines, treatment safety (risk ratios/pooled incidence rates) and neurological/functional efficacy (Hedge’s g) were all evaluated. Results: Twenty-three studies examined 17 treatments. Reporting quality scores were medium to high, but adherence to recommended guidelines was lower. Three treatments resulted in serious adverse events; four improved outcomes more than standard care. However, many studies were under-powered and individual patients varied in their response to some treatments. Conclusion: Preliminary findings suggest that some cell therapies may be relatively safe and effective, but larger double-blinded placebo-controlled studies are needed to establish the long-term risks and benefits.
Collapse
Affiliation(s)
- David J Unsworth
- Faculty of Health & Medical Sciences, University of Adelaide, Adelaide, South Australia, Australia
| | - Jane L Mathias
- Faculty of Health & Medical Sciences, University of Adelaide, Adelaide, South Australia, Australia
| | - Diana S Dorstyn
- Faculty of Health & Medical Sciences, University of Adelaide, Adelaide, South Australia, Australia
| |
Collapse
|
24
|
Yang R, Xu C, Wang T, Wang Y, Wang J, Quan D, Deng DYB. PTMAc-PEG-PTMAc hydrogel modified by RGDC and hyaluronic acid promotes neural stem cells' survival and differentiation in vitro. RSC Adv 2017. [DOI: 10.1039/c7ra06614g] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
The enhancement of the biological properties of hydrogels by surface modifying with bioactive molecules is of great significance, especially for the treatment of central nervous system injury by combining engrafted cells.
Collapse
Affiliation(s)
- Ruirui Yang
- Research Center of Translational Medicine
- The First Affiliated Hospital
- Sun Yat-sen University
- Guangzhou 510080
- China
| | - Caixia Xu
- Research Center of Translational Medicine
- The First Affiliated Hospital
- Sun Yat-sen University
- Guangzhou 510080
- China
| | - Tao Wang
- PCFM Lab
- GD HPPC Lab
- Research Center of Engineering and Technology for Functional Biomaterials of Guangdong
- School of Chemistry
- Sun Yat-Sen University
| | - Yuanqi Wang
- Research Center of Translational Medicine
- The First Affiliated Hospital
- Sun Yat-sen University
- Guangzhou 510080
- China
| | - Jingnan Wang
- Research Center of Translational Medicine
- The First Affiliated Hospital
- Sun Yat-sen University
- Guangzhou 510080
- China
| | - Daping Quan
- PCFM Lab
- GD HPPC Lab
- Research Center of Engineering and Technology for Functional Biomaterials of Guangdong
- School of Chemistry
- Sun Yat-Sen University
| | - David Y. B. Deng
- Research Center of Translational Medicine
- The First Affiliated Hospital
- Sun Yat-sen University
- Guangzhou 510080
- China
| |
Collapse
|
25
|
French BR, Boddepalli RS, Govindarajan R. Acute Ischemic Stroke: Current Status and Future Directions. MISSOURI MEDICINE 2016; 113:480-486. [PMID: 30228538 PMCID: PMC6139763] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
The evolving knowledge on stroke in conjunction with advances in the field of imaging, treatment approaches using recombinant tissue plasminogen activator (rtPA) or thrombectomy devices in recanalization, and efficient emergency stroke workflow processes have opened new frontiers in managing patients with an acute ischemic stroke. These frontiers have been reformed and overcome in overcoming the decades-long watch and wait approach towards patients with ischemic stroke. In this article, we focus on the current strategies for managing ischemic stroke and conclude by providing a brief overview of anticipating developments that can transform future stroke treatments.
Collapse
Affiliation(s)
- Brandi R French
- Brandi R. French, MD, Assistant Professor of Clinical Vascular Neurology, Medical Director of Inpatient Neurosciences Unit in the Department of Neurology, University of Missouri - Columbia, Missouri
| | - Raja S Boddepalli
- Raja S. Boddepalli, MD, Research Assistant in the Department of Neurology, University of Missouri - Columbia, Missouri
| | - Raghav Govindarajan
- Raghav Govindarajan MD, FISQua, FACSc, FCCP, MSMA member since 2013 and 2017 Boone County Medical society President, Assistant Professor in the Department of Neurology, University of Missouri - Columbia, Missouri
| |
Collapse
|
26
|
Ye Y, Peng YR, Hu SQ, Yan XL, Chen J, Xu T. In Vitro Differentiation of Bone Marrow Mesenchymal Stem Cells into Neuron-Like Cells by Cerebrospinal Fluid Improves Motor Function of Middle Cerebral Artery Occlusion Rats. Front Neurol 2016; 7:183. [PMID: 27833584 PMCID: PMC5081354 DOI: 10.3389/fneur.2016.00183] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2016] [Accepted: 10/10/2016] [Indexed: 11/13/2022] Open
Abstract
Bone marrow mesenchymal stem cells (BMSCs) represent a promising tool for stem cell-based therapies. However, the majority of BMSC transplants only allow for limited recovery of the lost functions. We previously found that human cerebrospinal fluid (hCSF) is more potent than growth factors in differentiating human BMSCs into neuron-like cells in vitro. In this study, we studied the effect of transplantation of rat BMSC-derived neuron-like cells (BMSC-Ns) induced by hCSF into rat brain with middle cerebral artery occlusion (MCAO). The survival and differentiation of the transplanted cells were determined using immunofluorescence staining of bromodeoxyuridine. The recovery of neurological function were observed by the modified neurological severity score (modified NSS) at 4, 15, and 32 days after cell transplantation, HE staining for determination of the infarct volume at day 32 after cell transplantation. Transplantation of BMSC-Ns or BMSCs significantly improved indexes of neurological function and reduced infarct size in rats previously subjected to MCAO compared with those in the control group. Remarkably, 32 days after transplantation, rats treated with BMSC-Ns presented a smaller infarct size, higher number of neuron-specific, enolase-positive, and BrdU-positive cells, and improved neurological function compared with BMSC group. Our results demonstrate that transplantation of hCSF-treated BMSC-Ns significantly improves neurological function and reduces infarct size in rats subjected to MCAO. This study may pave a new avenue for the treatment of MCAO.
Collapse
Affiliation(s)
- Ying Ye
- Jiangsu Province Key Laboratory of Anesthesiology, Institute of Emergency Rescue Medicine, Xuzhou Medical University, Xuzhou, Jiangsu, China; Emergency Center, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Yi-Ran Peng
- Department of Clinical Medicine, Xuzhou Medical University , Xuzhou , China
| | - Shu-Qun Hu
- Jiangsu Province Key Laboratory of Anesthesiology, Institute of Emergency Rescue Medicine, Xuzhou Medical University , Xuzhou, Jiangsu , China
| | - Xian-Liang Yan
- Jiangsu Province Key Laboratory of Anesthesiology, Institute of Emergency Rescue Medicine, Xuzhou Medical University, Xuzhou, Jiangsu, China; Emergency Center, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Juan Chen
- Jiangsu Province Key Laboratory of Anesthesiology, Institute of Emergency Rescue Medicine, Xuzhou Medical University , Xuzhou, Jiangsu , China
| | - Tie Xu
- Jiangsu Province Key Laboratory of Anesthesiology, Institute of Emergency Rescue Medicine, Xuzhou Medical University, Xuzhou, Jiangsu, China; Emergency Center, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| |
Collapse
|
27
|
Inhibition of HSP90 Promotes Neural Stem Cell Survival from Oxidative Stress through Attenuating NF- κB/p65 Activation. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2016; 2016:3507290. [PMID: 27818721 PMCID: PMC5080492 DOI: 10.1155/2016/3507290] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/08/2016] [Revised: 08/23/2016] [Accepted: 08/25/2016] [Indexed: 01/19/2023]
Abstract
Stem cell survival after transplantation determines the efficiency of stem cell treatment, which develops as a novel potential therapy for several central nervous system (CNS) diseases in recent decades. The engrafted stem cells face the damage of oxidative stress, inflammation, and immune response at the lesion point in host. Among the damaging pathologies, oxidative stress directs stem cells to apoptosis and even death through several signalling pathways and DNA damage. However, the in-detail mechanism of stem cell survival from oxidative stress has not been revealed clearly. Here, in this study, we used hydrogen peroxide (H2O2) to induce the oxidative damage on neural stem cells (NSCs). The damage was in consequence demonstrated involving the activation of heat shock protein 90 (HSP90) and NF-κB/p65 signalling pathways. Further application of the pharmacological inhibitors, respectively, targeting at each signalling indicated an upper-stream role of HSP90 upon NF-κB/p65 on NSCs survival. Preinhibition of HSP90 with the specific inhibitor displayed a significant protection on NSCs against oxidative stress. In conclusion, inhibition of HSP90 would attenuate NF-κB/p65 activation by oxidative induction and promote NSCs survival from oxidative damage. The HSP90/NF-κB mechanism provides a new evidence on rescuing NSCs from oxidative stress and also promotes the stem cell application on CNS pathologies.
Collapse
|
28
|
Van Pham P, Vu NB, Nguyen HT, Phan NK. Isolation of endothelial progenitor cells from human adipose tissue. BIOMEDICAL RESEARCH AND THERAPY 2016. [DOI: 10.7603/s40730-016-0024-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|