1
|
García Granado JF, Rodríguez Esparragón FJ, González Martín JM, Cazorla Rivero SE, González Hernández AN. Endothelial and circulating progenitor cells as prognostic biomarkers of stroke: A systematic review and meta-analysis. Thromb Res 2025; 245:109224. [PMID: 39571224 DOI: 10.1016/j.thromres.2024.109224] [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: 07/02/2024] [Revised: 10/19/2024] [Accepted: 11/15/2024] [Indexed: 12/30/2024]
Abstract
PURPOSE Endothelial progenitor cells (EPCs) are biomarkers of neurovascular repair in cerebral vascular disease (CVD). Low quantification of EPCs and/or their dysfunction has been associated with stroke severity and post-stroke functionality. This systematic review (SR) and meta-analysis aimed to analyze whether EPC quantification contributes to stroke severity and functional prognosis. METHODS Articles were selected from the PubMed, ScienceDirect, and Ovid MEDLINE databases, according to the guidelines of the PRISMA 2020 [1] statement. Detailed observational studies of samples from subjects with a clinical diagnosis of CVD (ischemic stroke-IS, hemorrhagic stroke-HS, or transient ischemic attack-TIA) aged >45 years during 2003-2023 were included. Evaluation of study quality was based on the Critical Appraisal Skills Programme checklist(Santamaría, 2017 [2]). RESULTS We included 22 articles in our SR. Patients with IS and good functional outcomes had higher EPC levels during the first week of admission than those with worse functional outcomes. Higher EPC levels were associated with reduced infarct growth, improved NIHSS scores at 48 h (OR 0.8; 95 % CI: 0.72-0.90; p < 0.0002) 7 (r = -0.607; p < 0.0001), and 90 days (r = -0.570; p < 0.0001), with a negative correlation between EPC levels and NIHSS score (overall pooled r = -0,32, 95 % CI: -0.39-0.24), and good functional outcomes with better mRS scores at 24 h, 3, 6, and 12 months (overall pooled SMD 4.51, CI 95 %: 0.70-0.83). Lower EPC quantification and worse functional outcomes during admission were predictors of IS recurrence. Higher EPC levels were associated with better functional outcomes and lower bleeding volumes in patients with HS and were protective markers for the progression high-risk TIA. CONCLUSION EPCs seems to be predictive biomarkers of better clinical outcomes in patients with CVD, exhibiting lower severity (NIHSS) and better functional prognosis (mRS).
Collapse
Affiliation(s)
- Juan Francisco García Granado
- Hospital Universitario de Gran Canaria Doctor Negrín, Universidad de Las Palmas de Gran Canaria (ULPGC), Las Palmas de Gran Canaria, Spain.
| | | | | | - Sara E Cazorla Rivero
- Hospital Universitario de Gran Canaria Doctor Negrín, Las Palmas de Gran Canaria, Spain
| | - Ayoze Nauzet González Hernández
- Hospital Universitario de Gran Canaria Doctor Negrín, Universidad Fernando Pessoa Canarias, Las Palmas de Gran Canaria, Spain
| |
Collapse
|
2
|
Penny TR, Jenkin G, Miller SL, McDonald CA. Umbilical cord blood derived cell expansion: a potential neuroprotective therapy. Stem Cell Res Ther 2024; 15:234. [PMID: 39075614 PMCID: PMC11287950 DOI: 10.1186/s13287-024-03830-0] [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: 01/07/2024] [Accepted: 07/02/2024] [Indexed: 07/31/2024] Open
Abstract
Umbilical cord blood (UCB) is a rich source of beneficial stem and progenitor cells with known angiogenic, neuroregenerative and immune-modulatory properties. Preclinical studies have highlighted the benefit of UCB for a broad range of conditions including haematological conditions, metabolic disorders and neurological conditions, however clinical translation of UCB therapies is lacking. One barrier for clinical translation is inadequate cell numbers in some samples meaning that often a therapeutic dose cannot be achieved. This is particularly important when treating adults or when administering repeat doses of cells. To overcome this, UCB cell expansion is being explored to increase cell numbers. The current focus of UCB cell expansion is CD34+ haematopoietic stem cells (HSCs) for which the main application is treatment of haematological conditions. Currently there are 36 registered clinical trials that are examining the efficacy of expanded UCB cells with 31 of these being for haematological malignancies. Early data from these trials suggest that expanded UCB cells are a safe and feasible treatment option and show greater engraftment potential than unexpanded UCB. Outside of the haematology research space, expanded UCB has been trialled as a therapy in only two preclinical studies, one for spinal cord injury and one for hind limb ischemia. Proteomic analysis of expanded UCB cells in these studies showed that the cells were neuroprotective, anti-inflammatory and angiogenic. These findings are also supported by in vitro studies where expanded UCB CD34+ cells showed increased gene expression of neurotrophic and angiogenic factors compared to unexpanded CD34+ cells. Preclinical evidence demonstrates that unexpanded CD34+ cells are a promising therapy for neurological conditions where they have been shown to improve multiple indices of injury in rodent models of stroke, Parkinson's disease and neonatal hypoxic ischemic brain injury. This review will highlight the current application of expanded UCB derived HSCs in transplant medicine, and also explore the potential use of expanded HSCs as a therapy for neurological conditions. It is proposed that expanded UCB derived CD34+ cells are an appropriate cellular therapy for a range of neurological conditions in children and adults.
Collapse
Affiliation(s)
- Tayla R Penny
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, VIC, Australia.
- Department of Obstetrics and Gynaecology, Monash University, Clayton, VIC, Australia.
| | - Graham Jenkin
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, VIC, Australia
- Department of Obstetrics and Gynaecology, Monash University, Clayton, VIC, Australia
| | - Suzanne L Miller
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, VIC, Australia
- Department of Obstetrics and Gynaecology, Monash University, Clayton, VIC, Australia
| | - Courtney A McDonald
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, VIC, Australia
- Department of Obstetrics and Gynaecology, Monash University, Clayton, VIC, Australia
| |
Collapse
|
3
|
Panos LD, Bargiotas P, Arnold M, Hadjigeorgiou G, Panos GD. Revolutionizing Stroke Recovery: Unveiling the Promise of Stem Cell Therapy. Drug Des Devel Ther 2024; 18:991-1006. [PMID: 38567255 PMCID: PMC10986404 DOI: 10.2147/dddt.s460998] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Accepted: 03/26/2024] [Indexed: 04/04/2024] Open
Abstract
Stem cells, renowned for their unique regenerative capabilities, present significant hope in treating stroke, a major cause of disability globally. This review offers a detailed analysis of stem cell applications in stroke (ischemic and hemorrhagic) recovery. It examines therapies based on autologous (patient-derived), allogeneic (donor-derived), and Granulocyte-Colony Stimulating Factor (G-CSF) based stem cells, focusing on cell types such as Mesenchymal Stem/Stromal Cells (MSCs), Bone Marrow Mononuclear Stem Cells (BMMSCs), and Neural Stem/Progenitor Cells (NSCs). The paper compiles clinical trial data to evaluate their effectiveness and safety and addresses the ethical concerns of these innovative treatments. By explaining the mechanisms of stem cell-induced neurological repair, this review underscores stem cells' potential in revolutionizing stroke rehabilitation and suggests avenues for future research.
Collapse
Affiliation(s)
- Leonidas D Panos
- Department of Neurology, Bern University Hospital Inselspital, Bern, Switzerland
- Department of Neurology, School of Medicine, University of Cyprus, Nicosia, Cyprus
| | - Panagiotis Bargiotas
- Department of Neurology, School of Medicine, University of Cyprus, Nicosia, Cyprus
| | - Marcel Arnold
- Department of Neurology, Bern University Hospital Inselspital, Bern, Switzerland
| | | | - Georgios D Panos
- Department of Ophthalmology, Queen’s Medical Centre, Nottingham University Hospitals (NUH), Nottingham, UK
- Division of Ophthalmology and Visual Sciences, School of Medicine, University of Nottingham, Nottingham, UK
| |
Collapse
|
4
|
Custodia A, Aramburu-Núñez M, Rodríguez-Arrizabalaga M, Pías-Peleteiro JM, Vázquez-Vázquez L, Camino-Castiñeiras J, Aldrey JM, Castillo J, Ouro A, Sobrino T, Romaus-Sanjurjo D. Biomarkers Assessing Endothelial Dysfunction in Alzheimer's Disease. Cells 2023; 12:cells12060962. [PMID: 36980302 PMCID: PMC10047803 DOI: 10.3390/cells12060962] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 03/07/2023] [Accepted: 03/10/2023] [Indexed: 03/30/2023] Open
Abstract
Alzheimer's disease (AD) is the most common degenerative disorder in the elderly in developed countries. Currently, growing evidence is pointing at endothelial dysfunction as a key player in the cognitive decline course of AD. As a main component of the blood-brain barrier (BBB), the dysfunction of endothelial cells driven by vascular risk factors associated with AD allows the passage of toxic substances to the cerebral parenchyma, producing chronic hypoperfusion that eventually causes an inflammatory and neurotoxic response. In this process, the levels of several biomarkers are disrupted, such as an increase in adhesion molecules that allow the passage of leukocytes to the cerebral parenchyma, increasing the permeability of the BBB; moreover, other vascular players, including endothelin-1, also mediate artery inflammation. As a consequence of the disruption of the BBB, a progressive neuroinflammatory response is produced that, added to the astrogliosis, eventually triggers neuronal degeneration (possibly responsible for cognitive deterioration). Recently, new molecules have been proposed as early biomarkers for endothelial dysfunction that can constitute new therapeutic targets as well as early diagnostic and prognostic markers for AD.
Collapse
Affiliation(s)
- Antía Custodia
- NeuroAging Group (NEURAL), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Marta Aramburu-Núñez
- NeuroAging Group (NEURAL), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Mariña Rodríguez-Arrizabalaga
- NeuroAging Group (NEURAL), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain
| | - Juan Manuel Pías-Peleteiro
- NeuroAging Group (NEURAL), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Laura Vázquez-Vázquez
- NeuroAging Group (NEURAL), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Javier Camino-Castiñeiras
- NeuroAging Group (NEURAL), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - José Manuel Aldrey
- NeuroAging Group (NEURAL), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - José Castillo
- Neuroimaging and Biotechnology Laboratory (NOBEL), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain
| | - Alberto Ouro
- NeuroAging Group (NEURAL), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Tomás Sobrino
- NeuroAging Group (NEURAL), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Daniel Romaus-Sanjurjo
- NeuroAging Group (NEURAL), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, 28029 Madrid, Spain
| |
Collapse
|
5
|
Yan S, Campos de Souza S, Xie Z, Bao Y. Research progress in clinical trials of stem cell therapy for stroke and neurodegenerative diseases. IBRAIN 2023; 9:214-230. [PMID: 37786546 PMCID: PMC10529019 DOI: 10.1002/ibra.12095] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Revised: 02/14/2023] [Accepted: 02/16/2023] [Indexed: 10/04/2023]
Abstract
The incidence of stroke and neurodegenerative diseases is gradually increasing in modern society, but there is still no treatment that is effective enough. Stem cells are cells that can reproduce (self-renew) and differentiate into the body, which have shown significance in basic research, while doctors have also taken them into clinical trials to determine their efficacy and safety. Existing clinical trials mainly include middle-aged and elderly patients with stroke or Parkinson's disease (mostly 40-80 years old), mainly involving injection of mesenchymal stem cells and bone marrow mesenchymal stem cells through the veins and the putamen, with a dosage of mostly 106-108 cells. The neural and motor functions of the patients were restored after stem cell therapy, and the safety was found to be good during the follow-up period of 3 months to 5 years. Here, we review all clinical trials and the latest advances in stroke, Alzheimer's disease, and Parkinson's disease, with the hope that stem cell therapy will be used in the clinic in the future to achieve effective treatment rates and benefit patients.
Collapse
Affiliation(s)
- Shan‐Shan Yan
- Department of AnesthesiologySouthwest Medical UniversityLuzhouChina
| | - Senio Campos de Souza
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical SciencesUniversity of MacauMacau SARChina
| | - Zhen‐Dong Xie
- Institute for Bioengineering of CataloniaUniversity of BarcelonaCarrer de Baldiri ReixacBarcelonaSpain
| | - Yong‐Xin Bao
- Qingdao Women and Children's HospitalQingdao UniversityQingdaoChina
| |
Collapse
|
6
|
Custodia A, Ouro A, Sargento-Freitas J, Aramburu-Núñez M, Pías-Peleteiro JM, Hervella P, Rosell A, Ferreira L, Castillo J, Romaus-Sanjurjo D, Sobrino T. Unraveling the potential of endothelial progenitor cells as a treatment following ischemic stroke. Front Neurol 2022; 13:940682. [PMID: 36158970 PMCID: PMC9492921 DOI: 10.3389/fneur.2022.940682] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Accepted: 08/15/2022] [Indexed: 11/13/2022] Open
Abstract
Ischemic stroke is becoming one of the most common causes of death and disability in developed countries. Since current therapeutic options are quite limited, focused on acute reperfusion therapies that are hampered by a very narrow therapeutic time window, it is essential to discover novel treatments that not only stop the progression of the ischemic cascade during the acute phase, but also improve the recovery of stroke patients during the sub-acute or chronic phase. In this regard, several studies have shown that endothelial progenitor cells (EPCs) can repair damaged vessels as well as generate new ones following cerebrovascular damage. EPCs are circulating cells with characteristics of both endothelial cells and adult stem cells presenting the ability to differentiate into mature endothelial cells and self-renew, respectively. Moreover, EPCs have the advantage of being already present in healthy conditions as circulating cells that participate in the maintenance of the endothelium in a direct and paracrine way. In this scenario, EPCs appear as a promising target to tackle stroke by self-promoting re-endothelization, angiogenesis and vasculogenesis. Based on clinical data showing a better neurological and functional outcome in ischemic stroke patients with higher levels of circulating EPCs, novel and promising therapeutic approaches would be pharmacological treatment promoting EPCs-generation as well as EPCs-based therapies. Here, we will review the latest advances in preclinical as well as clinical research on EPCs application following stroke, not only as a single treatment but also in combination with new therapeutic approaches.
Collapse
Affiliation(s)
- Antía Custodia
- NeuroAging Laboratory (NEURAL), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain
| | - Alberto Ouro
- NeuroAging Laboratory (NEURAL), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain
| | - João Sargento-Freitas
- Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal
- Faculdade de Medicina da Universidade de Coimbra, Coimbra, Portugal
- Centro Neurociências e Biologia Celular, Coimbra, Portugal
| | - Marta Aramburu-Núñez
- NeuroAging Laboratory (NEURAL), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain
| | - Juan Manuel Pías-Peleteiro
- NeuroAging Laboratory (NEURAL), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain
| | - Pablo Hervella
- Neuroimaging and Biotechnology Laboratory (NOBEL), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain
| | - Anna Rosell
- Neurovascular Research Laboratory, Vall d'Hebron Research Institute, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Lino Ferreira
- Faculdade de Medicina da Universidade de Coimbra, Coimbra, Portugal
- Centro Neurociências e Biologia Celular, Coimbra, Portugal
- CNC-Center for Neuroscience and Cell Biology, CIBB-Centre for Innovative Biomedicine and Biotechnology, UC, Biotech Parque Tecnológico de Cantanhede, University of Coimbra, Coimbra, Portugal
| | - José Castillo
- Neuroimaging and Biotechnology Laboratory (NOBEL), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain
| | - Daniel Romaus-Sanjurjo
- NeuroAging Laboratory (NEURAL), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain
- *Correspondence: Daniel Romaus-Sanjurjo
| | - Tomás Sobrino
- NeuroAging Laboratory (NEURAL), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain
- Tomás Sobrino
| |
Collapse
|
7
|
Custodia A, Ouro A, Romaus-Sanjurjo D, Pías-Peleteiro JM, de Vries HE, Castillo J, Sobrino T. Endothelial Progenitor Cells and Vascular Alterations in Alzheimer’s Disease. Front Aging Neurosci 2022; 13:811210. [PMID: 35153724 PMCID: PMC8825416 DOI: 10.3389/fnagi.2021.811210] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Accepted: 12/14/2021] [Indexed: 12/14/2022] Open
Abstract
Alzheimer’s disease (AD) is a neurodegenerative disease representing the most common type of dementia worldwide. The early diagnosis of AD is very difficult to achieve due to its complexity and the practically unknown etiology. Therefore, this is one of the greatest challenges in the field in order to develop an accurate therapy. Within the different etiological hypotheses proposed for AD, we will focus on the two-hit vascular hypothesis and vascular alterations occurring in the disease. According to this hypothesis, the accumulation of β-amyloid protein in the brain starts as a consequence of damage in the cerebral vasculature. Given that there are several vascular and angiogenic alterations in AD, and that endothelial progenitor cells (EPCs) play a key role in endothelial repair processes, the study of EPCs in AD may be relevant to the disease etiology and perhaps a biomarker and/or therapeutic target. This review focuses on the involvement of endothelial dysfunction in the onset and progression of AD with special emphasis on EPCs as a biomarker and potential therapeutic target.
Collapse
Affiliation(s)
- Antía Custodia
- NeuroAging Group (NEURAL), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain
| | - Alberto Ouro
- NeuroAging Group (NEURAL), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain
- *Correspondence: Alberto Ouro,
| | - Daniel Romaus-Sanjurjo
- NeuroAging Group (NEURAL), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain
| | - Juan Manuel Pías-Peleteiro
- NeuroAging Group (NEURAL), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain
| | - Helga E. de Vries
- Neuroimmunology Research Group, Department of Molecular Cell Biology and Immunology, Amsterdam Neuroscience, Amsterdam UMC, Amsterdam, Netherlands
| | - José Castillo
- Neuroimaging and Biotechnology Laboratory (NOBEL), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain
| | - Tomás Sobrino
- NeuroAging Group (NEURAL), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain
- Tomás Sobrino,
| |
Collapse
|
8
|
Endothelial Progenitor Cells as a Marker of Vascular Damage But not a Predictor in Acute Microangiopathy-Associated Stroke. J Clin Med 2020; 9:jcm9072248. [PMID: 32679827 PMCID: PMC7408782 DOI: 10.3390/jcm9072248] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Revised: 07/13/2020] [Accepted: 07/14/2020] [Indexed: 02/02/2023] Open
Abstract
Background: The aim of the study was to assess the number of endothelial progenitor cells (EPCs) in patients with acute stroke due to cerebral microangiopathy and evaluate whether there is a relationship between their number and clinical status, radiological findings, risk factors, selected biochemical parameters, and prognosis, both in ischemic and hemorrhagic stroke. Methods: In total, 66 patients with lacunar ischemic stroke, 38 patients with typical location hemorrhagic stroke, and 22 subjects from the control group without acute cerebrovascular incidents were included in the prospective observational study. The number of EPCs was determined in serum on the first and eighth day after stroke onset using flow cytometry and identified with the immune-phenotype classification determinant (CD)45−, CD34+, CD133+. Results: We demonstrated a significantly higher number of EPCs on the first day of stroke compared to the control group (med. 17.75 cells/µL (0–488 cells/µL) vs. 5.24 cells/µL (0–95 cells/µL); p = 0.0006). We did not find a relationship between the number of EPCs in the acute phase of stroke and the biochemical parameters, vascular risk factors, or clinical condition. In females, the higher number of EPCs on the first day of stroke is related to a favorable functional outcome on the eighth day after the stroke onset compared to males (p = 0.0355). We found that a higher volume of the hemorrhagic focus on the first day was correlated with a lower number of EPCs on the first day (correlation coefficient (R) = −0.3378, p = 0.0471), and a higher number of EPCs on the first day of the hemorrhagic stroke was correlated with a lower degree of regression of the hemorrhagic focus (R = −0.3896, p = 0.0367). Conclusion: The study showed that endothelial progenitor cells are an early marker in acute microangiopathy-associated stroke regardless of etiology and may affect the radiological findings in hemorrhagic stroke. Nevertheless, their prognostic value remains doubtful in stroke patients.
Collapse
|
9
|
Abdulkadir RR, Alwjwaj M, Othman OA, Rakkar K, Bayraktutan U. Outgrowth endothelial cells form a functional cerebral barrier and restore its integrity after damage. Neural Regen Res 2020; 15:1071-1078. [PMID: 31823887 PMCID: PMC7034270 DOI: 10.4103/1673-5374.269029] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Breakdown of blood-brain barrier, formed mainly by brain microvascular endothelial cells (BMECs), represents the major cause of mortality during early phases of ischemic strokes. Hence, discovery of novel agents that can effectively replace dead or dying endothelial cells to restore blood-brain barrier integrity is of paramount importance in stroke medicine. Although endothelial progenitor cells (EPCs) represent one such agents, their rarity in peripheral blood severely limits their adequate isolation and therapeutic use for acute ischemic stroke which necessitate their ex vivo expansion and generate early EPCs and outgrowth endothelial cells (OECs) as a result. Functional analyses of these cells, in the present study, demonstrated that only OECs endocytosed DiI-labelled acetylated low-density lipoprotein and formed tubules on matrigel, prominent endothelial cell and angiogenesis markers, respectively. Further analyses by flow cytometry demonstrated that OECs expressed specific markers for stemness (CD34), immaturity (CD133) and endothelial cells (CD31) but not for hematopoietic cells (CD45). Like BMECs, OECs established an equally tight in vitro model of human BBB with astrocytes and pericytes, suggesting their capacity to form tight junctions. Ischemic injury mimicked by concurrent deprivation of oxygen and glucose (4 hours) or deprivation of oxygen and glucose followed by reperfusion (20 hours) affected both barrier integrity and function in a similar fashion as evidenced by decreases in transendothelial electrical resistance and increases in paracellular flux, respectively. Wound scratch assays comparing the vasculoreparative capacity of cells revealed that, compared to BMECs, OECs possessed a greater proliferative and directional migratory capacity. In a triple culture model of BBB established with astrocytes, pericytes and BMEC, exogenous addition of OECs effectively repaired the damage induced on endothelial layer in serum-free conditions. Taken together, these data demonstrate that OECs may effectively home to the site of vascular injury and repair the damage to maintain (neuro)vascular homeostasis during or after a cerebral ischemic injury.
Collapse
Affiliation(s)
- Rais Reskiawan Abdulkadir
- Stroke, Division of Clinical Neuroscience, Clinical Sciences Building, School of Medicine, Hucknall Road, Nottingham, NG5 1PB, UK
| | - Mansour Alwjwaj
- Stroke, Division of Clinical Neuroscience, Clinical Sciences Building, School of Medicine, Hucknall Road, Nottingham, NG5 1PB, UK
| | - Othman Ahmad Othman
- Stroke, Division of Clinical Neuroscience, Clinical Sciences Building, School of Medicine, Hucknall Road, Nottingham, NG5 1PB, UK
| | - Kamini Rakkar
- Stroke, Division of Clinical Neuroscience, Clinical Sciences Building, School of Medicine, Hucknall Road, Nottingham, NG5 1PB, UK
| | - Ulvi Bayraktutan
- Stroke, Division of Clinical Neuroscience, Clinical Sciences Building, School of Medicine, Hucknall Road, Nottingham, NG5 1PB, UK
- Correspondence to: Ulvi Bayraktutan, .
| |
Collapse
|
10
|
Fernández-Susavila H, Bugallo-Casal A, Castillo J, Campos F. Adult Stem Cells and Induced Pluripotent Stem Cells for Stroke Treatment. Front Neurol 2019; 10:908. [PMID: 31555195 PMCID: PMC6722184 DOI: 10.3389/fneur.2019.00908] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2019] [Accepted: 08/05/2019] [Indexed: 12/14/2022] Open
Abstract
Stroke is the main cause of disability and death in the world within neurological diseases. Despite such a huge impact, enzymatic, and mechanical recanalization are the only treatments available so far for ischemic stroke, but only <20% of patients can benefit from them. The use of stem cells as a possible cell therapy in stroke has been tested for years. The results obtained from these studies, although conflicting or controversial in some aspects, are promising. In the last few years, the recent development of the induced pluripotent stem cells has opened new possibilities to find new cell therapies against stroke. In this review, we will provide an overview of the state of the art of cell therapy in stroke. We will describe the current situation of the most employed stem cells and the use of induced pluripotent stem cells in stroke pathology. We will also present a summary of the different clinical trials that are being carried out or that already have results on the use of stem cells as a potential therapeutic intervention for stroke.
Collapse
Affiliation(s)
- Héctor Fernández-Susavila
- Clinical Neuroscience Research Laboratory, Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain
| | - Ana Bugallo-Casal
- Clinical Neuroscience Research Laboratory, Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain
| | - José Castillo
- Clinical Neuroscience Research Laboratory, Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain
| | - Francisco Campos
- Clinical Neuroscience Research Laboratory, Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain
| |
Collapse
|
11
|
Endothelial progenitor cells: Potential novel therapeutics for ischaemic stroke. Pharmacol Res 2019; 144:181-191. [DOI: 10.1016/j.phrs.2019.04.017] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Revised: 04/08/2019] [Accepted: 04/16/2019] [Indexed: 01/15/2023]
|
12
|
Liem NT, Huyen TL, Huong LT, Doan NV, Anh BV, Anh NTP, Tung DT. Outcomes of Bone Marrow Mononuclear Cell Transplantation for Neurological Sequelae Due to Intracranial Hemorrhage Incidence in the Neonatal Period: Report of Four Cases. Front Pediatr 2019; 7:543. [PMID: 32039110 PMCID: PMC6993568 DOI: 10.3389/fped.2019.00543] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Accepted: 12/12/2019] [Indexed: 12/24/2022] Open
Abstract
Aim: The aim of this study was to present primary outcomes of autologous bone marrow mononuclear cell (BMMNC) transplantation to improve neurological sequelae in four children with intracranial hemorrhage (ICH) incidence during the neonatal period. Methods: GMFM88 and modified Ashworth score were used to assess motor function and muscle spasticity before BMMNC transplantation and after transplantation. Brain MRI was performed to evaluate brain morphology before and after BMMNC transplantation. Bone marrow were harvested from anterior iliac crest puncture and BMMNCs were isolated using Ficoll gradient centrifugation. The microbiological testing, cell counting, and hematopoietic stem cell (hHSC CD34+ cell) analysis were performed, following which BMMNCs were infused intrathecally. Results: Improvement in motor function was observed in all patients after transplantation. In addition, muscle spasticity was reduced in all four patients. Conclusion: Autologous BMMNC transplantation may improve motor function and reduce muscle spasticity in children with ICH incidence during the neonatal period.
Collapse
Affiliation(s)
- Nguyen Thanh Liem
- Cellular Manufacturing Department, Vinmec Research Institute of Stem Cell and Gene Technology, Hanoi, Vietnam
| | - Truong Linh Huyen
- Cellular Manufacturing Department, Vinmec Research Institute of Stem Cell and Gene Technology, Hanoi, Vietnam
| | - Le Thu Huong
- Vinmec Times City General Hospital, Hanoi, Vietnam
| | - Ngo Van Doan
- Vinmec Times City General Hospital, Hanoi, Vietnam
| | - Bui Viet Anh
- Cellular Manufacturing Department, Vinmec Research Institute of Stem Cell and Gene Technology, Hanoi, Vietnam
| | | | | |
Collapse
|
13
|
Gao L, Xu W, Li T, Chen J, Shao A, Yan F, Chen G. Stem Cell Therapy: A Promising Therapeutic Method for Intracerebral Hemorrhage. Cell Transplant 2018; 27:1809-1824. [PMID: 29871521 PMCID: PMC6300771 DOI: 10.1177/0963689718773363] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Revised: 03/09/2018] [Accepted: 04/02/2018] [Indexed: 12/28/2022] Open
Abstract
Spontaneous intracerebral hemorrhage (ICH) is one type of the most devastating cerebrovascular diseases worldwide, which causes high morbidity and mortality. However, efficient treatment is still lacking. Stem cell therapy has shown good neuroprotective and neurorestorative effect in ICH and is a promising treatment. In this study, our aim was to review the therapeutic effects, strategies, related mechanisms and safety issues of various types of stem cell for ICH treatment. Numerous studies had demonstrated the therapeutic effects of diverse stem cell types in ICH. The potential mechanisms include tissue repair and replacement, neurotrophy, promotion of neurogenesis and angiogenesis, anti-apoptosis, immunoregulation and anti-inflammation and so forth. The microenvironment of the central nervous system (CNS) can also influence the effects of stem cell therapy. The detailed therapeutic strategies for ICH treatment such as cell type, the number of cells, time window, and the routes of medication delivery, varied greatly among different studies and had not been determined. Moreover, the safety issues of stem cell therapy for ICH should not be ignored. Stem cell therapy showed good therapeutic effect in ICH, making it a promising treatment. However, safety should be carefully evaluated, and more clinical trials are required before stem cell therapy can be extensively applied to clinical use.
Collapse
Affiliation(s)
- Liansheng Gao
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Zhejiang, China
| | - Weilin Xu
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Zhejiang, China
| | - Tao Li
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Zhejiang, China
| | - Jingyin Chen
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Zhejiang, China
| | - Anwen Shao
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Zhejiang, China
| | - Feng Yan
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Zhejiang, China
| | - Gao Chen
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Zhejiang, China
| |
Collapse
|
14
|
Johannesen S, Budeus B, Peters S, Iberl S, Meyer AL, Kammermaier T, Wirkert E, Bruun TH, Samara VC, Schulte-Mattler W, Herr W, Schneider A, Grassinger J, Bogdahn U. Biomarker Supervised G-CSF (Filgrastim) Response in ALS Patients. Front Neurol 2018; 9:971. [PMID: 30534107 PMCID: PMC6275232 DOI: 10.3389/fneur.2018.00971] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Accepted: 10/29/2018] [Indexed: 01/16/2023] Open
Abstract
Objective: To evaluate safety, tolerability and feasibility of long-term treatment with Granulocyte-colony stimulating factor (G-CSF), a well-known hematopoietic stem cell factor, guided by assessment of mobilized bone marrow derived stem cells and cytokines in the serum of patients with amyotrophic lateral sclerosis (ALS) treated on a named patient basis. Methods: 36 ALS patients were treated with subcutaneous injections of G-CSF on a named patient basis and in an outpatient setting. Drug was dosed by individual application schemes (mean 464 Mio IU/month, range 90-2160 Mio IU/month) over a median of 13.7 months (range from 2.7 to 73.8 months). Safety, tolerability, survival and change in ALSFRS-R were observed. Hematopoietic stem cells were monitored by flow cytometry analysis of circulating CD34+ and CD34+CD38− cells, and peripheral cytokines were assessed by electrochemoluminescence throughout the intervention period. Analysis of immunological and hematological markers was conducted. Results: Long term and individually adapted treatment with G-CSF was well tolerated and safe. G-CSF led to a significant mobilization of hematopoietic stem cells into the peripheral blood. Higher mobilization capacity was associated with prolonged survival. Initial levels of serum cytokines, such as MDC, TNF-beta, IL-7, IL-16, and Tie-2 were significantly associated with survival. Continued application of G-CSF led to persistent alterations in serum cytokines and ongoing measurements revealed the multifaceted effects of G-CSF. Conclusions: G-CSF treatment is feasible and safe for ALS patients. It may exert its beneficial effects through neuroprotective and -regenerative activities, mobilization of hematopoietic stem cells and regulation of pro- and anti-inflammatory cytokines as well as angiogenic factors. These cytokines may serve as prognostic markers when measured at the time of diagnosis. Hematopoietic stem cell numbers and cytokine levels are altered by ongoing G-CSF application and may potentially serve as treatment biomarkers for early monitoring of G-CSF treatment efficacy in ALS in future clinical trials.
Collapse
Affiliation(s)
- Siw Johannesen
- Department of Neurology, University Hospital Regensburg, Regensburg, Germany
| | | | - Sebastian Peters
- Department of Neurology, University Hospital Regensburg, Regensburg, Germany
| | - Sabine Iberl
- Department of Hematology, Internal Medicine III, University Hospital Regensburg, Regensburg, Germany
| | - Anne-Louise Meyer
- Department of Neurology, University Hospital Regensburg, Regensburg, Germany
| | - Tina Kammermaier
- Department of Neurology, University Hospital Regensburg, Regensburg, Germany
| | - Eva Wirkert
- Department of Neurology, University Hospital Regensburg, Regensburg, Germany
| | - Tim-Henrik Bruun
- Department of Neurology, University Hospital Regensburg, Regensburg, Germany
| | - Verena C Samara
- Stanford Neuroscience Health Center, Palo Alto, CA, United States
| | | | - Wolfgang Herr
- Department of Hematology, Internal Medicine III, University Hospital Regensburg, Regensburg, Germany
| | | | - Jochen Grassinger
- Department of Hematology, Internal Medicine III, University Hospital Regensburg, Regensburg, Germany
| | - Ulrich Bogdahn
- Department of Neurology, University Hospital Regensburg, Regensburg, Germany
| |
Collapse
|
15
|
Da Silva-Candal A, Argibay B, Iglesias-Rey R, Vargas Z, Vieites-Prado A, López-Arias E, Rodríguez-Castro E, López-Dequidt I, Rodríguez-Yáñez M, Piñeiro Y, Sobrino T, Campos F, Rivas J, Castillo J. Vectorized nanodelivery systems for ischemic stroke: a concept and a need. J Nanobiotechnology 2017; 15:30. [PMID: 28399863 PMCID: PMC5387212 DOI: 10.1186/s12951-017-0264-7] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2017] [Accepted: 04/03/2017] [Indexed: 02/07/2023] Open
Abstract
Neurological diseases of diverse aetiologies have significant effects on the quality of life of patients. The limited self-repairing capacity of the brain is considered to be the origin of the irreversible and progressive nature of many neurological diseases. Therefore, neuroprotection is an important goal shared by many clinical neurologists and neuroscientists. In this review, we discuss the main obstacles that have prevented the implementation of experimental neuroprotective strategies in humans and propose alternative avenues for the use of neuroprotection as a feasible therapeutic approach. Special attention is devoted to nanotechnology, which is a new approach for developing highly specific and localized biomedical solutions for the study of the multiple mechanisms involved in stroke. Nanotechnology is contributing to personalized neuroprotection by allowing us to identify mechanisms, determine optimal therapeutic windows, and protect patients from brain damage. In summary, multiple aspects of these new players in biomedicine should be considered in future in vivo and in vitro studies with the aim of improving their applicability to clinical studies.
Collapse
Affiliation(s)
- Andrés Da Silva-Candal
- Department of Neurology, Clinical Neurosciences Research Laboratory, Hospital Clínico Universitario, Universidade de Santiago de Compostela, Health Research Institute of Santiago de Compostela (IDIS), c/Travesa da Choupana, s/n, 15706, Santiago de Compostela, Spain
| | - Bárbara Argibay
- Department of Neurology, Clinical Neurosciences Research Laboratory, Hospital Clínico Universitario, Universidade de Santiago de Compostela, Health Research Institute of Santiago de Compostela (IDIS), c/Travesa da Choupana, s/n, 15706, Santiago de Compostela, Spain
| | - Ramón Iglesias-Rey
- Department of Neurology, Clinical Neurosciences Research Laboratory, Hospital Clínico Universitario, Universidade de Santiago de Compostela, Health Research Institute of Santiago de Compostela (IDIS), c/Travesa da Choupana, s/n, 15706, Santiago de Compostela, Spain
| | - Zulema Vargas
- Nanomag Laboratory, Department of Applied Physics, Technological Research Institute, Universidade de Santiago de Compostela, Health Research Institute of Santiago de Compostela (IDIS), Campus Vida, 15782, Santiago de Compostela, Spain
| | - Alba Vieites-Prado
- Department of Neurology, Clinical Neurosciences Research Laboratory, Hospital Clínico Universitario, Universidade de Santiago de Compostela, Health Research Institute of Santiago de Compostela (IDIS), c/Travesa da Choupana, s/n, 15706, Santiago de Compostela, Spain
| | - Esteban López-Arias
- Department of Neurology, Clinical Neurosciences Research Laboratory, Hospital Clínico Universitario, Universidade de Santiago de Compostela, Health Research Institute of Santiago de Compostela (IDIS), c/Travesa da Choupana, s/n, 15706, Santiago de Compostela, Spain
| | - Emilio Rodríguez-Castro
- Department of Neurology, Clinical Neurosciences Research Laboratory, Hospital Clínico Universitario, Universidade de Santiago de Compostela, Health Research Institute of Santiago de Compostela (IDIS), c/Travesa da Choupana, s/n, 15706, Santiago de Compostela, Spain
| | - Iria López-Dequidt
- Department of Neurology, Clinical Neurosciences Research Laboratory, Hospital Clínico Universitario, Universidade de Santiago de Compostela, Health Research Institute of Santiago de Compostela (IDIS), c/Travesa da Choupana, s/n, 15706, Santiago de Compostela, Spain
| | - Manuel Rodríguez-Yáñez
- Department of Neurology, Clinical Neurosciences Research Laboratory, Hospital Clínico Universitario, Universidade de Santiago de Compostela, Health Research Institute of Santiago de Compostela (IDIS), c/Travesa da Choupana, s/n, 15706, Santiago de Compostela, Spain
| | - Yolanda Piñeiro
- Nanomag Laboratory, Department of Applied Physics, Technological Research Institute, Universidade de Santiago de Compostela, Health Research Institute of Santiago de Compostela (IDIS), Campus Vida, 15782, Santiago de Compostela, Spain
| | - Tomás Sobrino
- Department of Neurology, Clinical Neurosciences Research Laboratory, Hospital Clínico Universitario, Universidade de Santiago de Compostela, Health Research Institute of Santiago de Compostela (IDIS), c/Travesa da Choupana, s/n, 15706, Santiago de Compostela, Spain
| | - Francisco Campos
- Department of Neurology, Clinical Neurosciences Research Laboratory, Hospital Clínico Universitario, Universidade de Santiago de Compostela, Health Research Institute of Santiago de Compostela (IDIS), c/Travesa da Choupana, s/n, 15706, Santiago de Compostela, Spain
| | - José Rivas
- Nanomag Laboratory, Department of Applied Physics, Technological Research Institute, Universidade de Santiago de Compostela, Health Research Institute of Santiago de Compostela (IDIS), Campus Vida, 15782, Santiago de Compostela, Spain.
| | - José Castillo
- Department of Neurology, Clinical Neurosciences Research Laboratory, Hospital Clínico Universitario, Universidade de Santiago de Compostela, Health Research Institute of Santiago de Compostela (IDIS), c/Travesa da Choupana, s/n, 15706, Santiago de Compostela, Spain.
| |
Collapse
|
16
|
Wang Y, Ji X, Leak RK, Chen F, Cao G. Stem cell therapies in age-related neurodegenerative diseases and stroke. Ageing Res Rev 2017; 34:39-50. [PMID: 27876573 PMCID: PMC5250574 DOI: 10.1016/j.arr.2016.11.002] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2016] [Revised: 09/26/2016] [Accepted: 11/04/2016] [Indexed: 02/06/2023]
Abstract
Aging, a complex process associated with various structural, functional and metabolic changes in the brain, is an important risk factor for neurodegenerative diseases and stroke. These diseases share similar neuropathological changes, such as the formation of misfolded proteins, oxidative stress, loss of neurons and synapses, dysfunction of the neurovascular unit (NVU), reduction of self-repair capacity, and motor and/or cognitive deficiencies. In addition to gray matter dysfunction, the plasticity and repair capacity of white matter also decrease with aging and contribute to neurodegenerative diseases. Aging not only renders patients more susceptible to these disorders, but also attenuates their self-repair capabilities. In addition, low drug responsiveness and intolerable side effects are major challenges in the prevention and treatment of senile diseases. Thus, stem cell therapies-characterized by cellular plasticity and the ability to self-renew-may be a promising strategy for aging-related brain disorders. Here, we review the common pathophysiological changes, treatments, and the promises and limitations of stem cell therapies in age-related neurodegenerative diseases and stroke.
Collapse
Affiliation(s)
- Yuan Wang
- Departments of Neurology, Xuanwu Hospital, Capital University of Medicine, Beijing 100053, China
| | - Xunming Ji
- Departments of Neurosurgery, Xuanwu Hospital, Capital University of Medicine, Beijing 100053, China
| | - Rehana K Leak
- Division of Pharmaceutical Sciences, Duquesne University, Pittsburgh, PA 15282, United States
| | - Fenghua Chen
- Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15260, United States
| | - Guodong Cao
- Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15260, United States; Geriatric Research Education and Clinical Centers, VA Pittsburgh Healthcare System, Pittsburgh, PA 15240, United States.
| |
Collapse
|
17
|
Pías-Peleteiro J, Campos F, Castillo J, Sobrino T. Endothelial progenitor cells as a therapeutic option in intracerebral hemorrhage. Neural Regen Res 2017; 12:558-561. [PMID: 28553326 PMCID: PMC5436344 DOI: 10.4103/1673-5374.205085] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Intracerebral hemorrhage (ICH) is the most severe cerebrovascular disease, which represents a leading cause of death and disability in developed countries. However, therapeutic options are limited, so is mandatory to investigate repairing processes after stroke in order to develop new therapeutic strategies able to promote brain repair processes. Therapeutic angiogenesis and vasculogenesis hold promise to improve outcome of ICH patients. In this regard, circulating endothelial progenitor cells (EPCs) have recently been suggested to be a marker of vascular risk and endothelial function. Moreover, EPC levels have been associated with good neurological and functional outcome as well as reduced residual hematoma volume in ICH patients. Finally, experimental and clinical studies indicate that EPC might mediate endothelial cell regeneration and neovascularization. Therefore, EPC-based therapy could be an excellent therapeutic option in ICH. In this mini-review, we discuss the present status of knowledge about the possible therapeutic role of EPCs in ICH, molecular mechanisms, and the future perspectives and strategies for their use in clinical practice.
Collapse
Affiliation(s)
- Juan Pías-Peleteiro
- Clinical Neurosciences Research Laboratory, Department of Neurology, Stroke Unit, University Clinical Hospital, Universidade de Santiago de Compostela, Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain
| | - Francisco Campos
- Clinical Neurosciences Research Laboratory, Department of Neurology, Stroke Unit, University Clinical Hospital, Universidade de Santiago de Compostela, Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain
| | - José Castillo
- Clinical Neurosciences Research Laboratory, Department of Neurology, Stroke Unit, University Clinical Hospital, Universidade de Santiago de Compostela, Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain
| | - Tomás Sobrino
- Clinical Neurosciences Research Laboratory, Department of Neurology, Stroke Unit, University Clinical Hospital, Universidade de Santiago de Compostela, Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain
| |
Collapse
|
18
|
Neovascularization and functional recovery after intracerebral hemorrhage is conditioned by the Tp53 Arg72Pro single-nucleotide polymorphism. Cell Death Differ 2016; 24:144-154. [PMID: 27768124 PMCID: PMC5260494 DOI: 10.1038/cdd.2016.109] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2016] [Revised: 08/29/2016] [Accepted: 09/07/2016] [Indexed: 02/06/2023] Open
Abstract
Intracerebral hemorrhage (ICH) is a devastating subtype of stroke that lacks effective therapy and reliable prognosis. Neovascularization following ICH is an essential compensatory response that mediates brain repair and modulates the clinical outcome of stroke patients. However, the mechanism that dictates this process is unknown. Bone marrow-derived endothelial progenitor cells (EPCs) promote endothelial repair and contribute to ischemia-induced neovascularization. The human Tp53 gene harbors a common single-nucleotide polymorphism (SNP) at codon 72, which yields an arginine-to-proline amino-acidic substitution (Arg72Pro) that modulates the apoptotic activity of the p53 protein. Previously, we found that this SNP controls neuronal susceptibility to ischemia-induced apoptosis in vitro. Here, we evaluated the impact of the Tp53 Arg72Pro SNP on vascular repair and functional recovery after ICH. We first analyzed EPC mobilization and functional outcome based on the modified Rankin scale scores in a hospital-based cohort of 78 patients with non-traumatic ICH. Patients harboring the Pro allele of the Tp53 Arg72Pro SNP showed higher levels of circulating EPC-containing CD34+ cells, EPC-mobilizing cytokines - vascular endothelial growth factor and stromal cell-derived factor-1α - and good functional outcome following ICH, when compared with the homozygous Arg allele patients, which is compatible with increased neovascularization. To assess directly whether Tp53 Arg72Pro SNP regulated neovascularization after ICH, we used the humanized Tp53 Arg72Pro knock-in mice, which were subjected to the collagenase-induced ICH. The brain endothelial cells of the Pro allele-carrying mice were highly resistant to ICH-mediated apoptosis, which facilitated cytokine-mediated EPC mobilization, cerebrovascular repair and functional recovery. However, these processes were not observed in the Arg allele-carrying mice. These results reveal that the Tp53 Arg72Pro SNP determines neovascularization, brain repair and neurological recovery after ICH. This study is the first in which the Pro allele of Tp53 is linked to vascular repair and ability to functionally recover from stroke.
Collapse
|
19
|
Increased Endothelial Progenitor Cell Levels are Associated with Good Outcome in Intracerebral Hemorrhage. Sci Rep 2016; 6:28724. [PMID: 27346699 PMCID: PMC4921860 DOI: 10.1038/srep28724] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2015] [Accepted: 06/08/2016] [Indexed: 01/25/2023] Open
Abstract
Circulating endothelial progenitor cells (EPCs) play a role in the regeneration of damaged brain tissue. However, the relationship between circulating EPC levels and functional recovery in intracerebral hemorrhage (ICH) has not yet been tested. Therefore, our aim was to study the influence of circulating EPCs on the outcome of ICH. Forty-six patients with primary ICH (males, 71.7%; age, 72.7 ± 10.8 years) were prospectively included in the study within 12 hours of symptom onset. The main outcome variable was good functional outcome at 12 months (modified Rankin scale ≤2), considering residual volume at 6 months as a secondary variable. Circulating EPC (CD34+/CD133+/KDR+) levels were measured by flow cytometry from blood samples obtained at admission, 72 hours and day 7. Our results indicate that patients with good outcome show higher EPC numbers at 72 hours and day 7 (all p < 0.001). However, only EPC levels at day 7 were independently associated with good functional outcome at 12 months (OR, 1.15; CI95%, 1.01–1.35) after adjustment by age, baseline stroke severity and ICH volume. Moreover, EPC levels at day 7 were negatively correlated to residual volume (r = −0.525; p = 0.005). In conclusion, these findings suggest that EPCs may play a role in the functional recovery of ICH patients.
Collapse
|
20
|
Senn R, Elkind MSV, Montaner J, Christ-Crain M, Katan M. Potential role of blood biomarkers in the management of nontraumatic intracerebral hemorrhage. Cerebrovasc Dis 2014; 38:395-409. [PMID: 25471997 DOI: 10.1159/000366470] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2014] [Accepted: 08/06/2014] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Intracerebral hemorrhage (ICH), a subtype of stroke associated with high mortality and disability, accounts for 13% of all strokes. Basic and clinical research has contributed to our understanding of the complex pathophysiology of neuronal injury in ICH. Outcome rates, however, remain stable, and questions regarding acute management of ICH remain unanswered. Newer research is aiming at matching measured levels of serum proteins, enzymes, or cells to different stages of brain damage, suggesting that blood biomarkers may assist in acute diagnosis, therapeutic decisions, and prognostication. This paper provides an overview on the most promising blood biomarkers and their potential role in the diagnosis and management of spontaneous ICH. SUMMARY Information was collected from studies, reviews, and guidelines listed in PubMed up to November 2013 on blood biomarkers of nontraumatic ICH in humans. We describe the potential role and limitations of GFAP, S100B/RAGE, and ApoC-III as diagnostic biomarkers, β-Amyloid as a biomarker for etiological classification, and 27 biomarkers for prognosis of mortality and functional outcome. Within the group of prognostic markers we discuss markers involved in coagulation processes (e.g., D-Dimers), neuroendocrine markers (e.g., copeptin), systemic metabolic markers (e.g., blood glucose levels), markers of inflammation (e.g., IL-6), as well as growth factors (e.g., VEGF), and others (e.g., glutamate). Some of those blood biomarkers are agents of pathologic processes associated with hemorrhagic stroke but also other diseases, whereas others play more distinct pathophysiological roles and help in understanding the basic mechanisms of brain damage and/or recovery in ICH. KEY MESSAGES Numerous blood biomarkers are associated with different pathophysiological pathways in ICH, and some of them promise to be useful in the management of ICH, eventually contributing additional information to current tools for diagnosis, therapy monitoring, risk stratification, or intervention. Up to date, however, no blood biomarker of ICH has been studied sufficiently to find its way into clinical routine yet; well-designed, large-scale, clinical studies addressing relevant clinical questions are needed. We suggest that the effectiveness of biomarker research in ICH might be improved by international cooperation and shared resources for large validation studies, such as provided by the consortium on stroke biomarker research (http://stroke-biomarkers.com/page.php?title=Resources).
Collapse
Affiliation(s)
- Rebecca Senn
- Department of Endocrinology, University Hospital Basel, Basel, Switzerland
| | | | | | | | | |
Collapse
|
21
|
Prakash A, Medhi B, Chopra K. Granulocyte colony stimulating factor (GCSF) improves memory and neurobehavior in an amyloid-β induced experimental model of Alzheimer's disease. Pharmacol Biochem Behav 2013; 110:46-57. [PMID: 23756182 DOI: 10.1016/j.pbb.2013.05.015] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2012] [Revised: 05/21/2013] [Accepted: 05/25/2013] [Indexed: 10/26/2022]
Abstract
GCSF is an endogenous neuronal hematopoietic factor that displays robust in vitro and in vivo neuroprotective activity. The present study aimed to evaluate the effect of GCSF on Aβ-induced memory loss in an Alzheimer's disease model of rats. A total of 42 male adult Wistar rats weighing 200-250 g were used in the study and were divided into 7 experimental groups. Animals were subjected to intracerebroventricular (ICV) injection stereotaxically at day 0 to instill amyloid-β(1-42) (Aβ(1-42)) or PBS (sham operated group) at 10 μl (5 μl bilaterally). GCSF treatment was given from day 7 to 12 of Aβ injection. On day 21, behavioral tests (short term memory, exploratory behavior and motor coordination) in all groups were evaluated. Biochemical parameters and RNA expression were measured to ensure the efficacy of GCSF. GCSF (35 and 70 μg/kg, s.c.) showed statistically significant improvement in memory as compared to control and sham operated groups (p<0.05). Mean time spent in the platform placed quadrant was found to be significantly increased in the GCSF (70 μg/kg, s.c.) as compared to GCSF (35 μg/kg, s.c.) and GCSF (10 μg/kg, s.c.) groups (p<0.001). GCSF (35 and 70 μg/kg, s.c.) also improved motor coordination and exploratory behavior significantly as compared to naïve sham operated and GCSF (10 μg/kg, s.c.) groups (p<0.05). Improvement in memory by GCSF (35 and 70 μg/kg, s.c.) was coupled with marked reduction of lipid peroxidation, acetylcholinesterase levels and a significant increase in antioxidant enzymes as well as total RNA expression in the brain. Additionally, GCSF (35 and 70 μg/kg, s.c.) significantly increased progenitor cells (iPSCs) and surface marker CD34+ in the brain and hence induced neurogenesis. The present findings demonstrate an improvement of memory and neurobehavioral function with GCSF in Aβ-induced Alzheimer's disease model in rats.
Collapse
Affiliation(s)
- Ajay Prakash
- Pharmacology Division, University Institute of Pharmaceutical Sciences, Panjab University, Chandigarh, India
| | | | | |
Collapse
|
22
|
Sobrino T, Pérez-Mato M, Brea D, Rodríguez-Yáñez M, Blanco M, Castillo J. Temporal profile of molecular signatures associated with circulating endothelial progenitor cells in human ischemic stroke. J Neurosci Res 2012; 90:1788-93. [PMID: 22513751 DOI: 10.1002/jnr.23068] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2011] [Revised: 03/07/2012] [Accepted: 03/20/2012] [Indexed: 11/11/2022]
Abstract
Endothelial progenitor cells (EPC) have been associated with good functional outcome in ischemic stroke. From preclinical studies, it has been reported that EPC proliferation is mediated by several molecular markers, including vascular endothelial growth factor (VEGF), stromal cell-derived factor-1α (SDF-1α), and the activity of matrix metalloproteinase-9 (MMP-9). Therefore, our aim was to study the role of these molecular factors in EPC proliferation in human ischemic stroke. Forty-eight patients with first episode of nonlacunar ischemic stroke were prospectively included in the study within 12 hr of symptom onset. EPC colonies were classified as early-outgrowth colony forming unit-endothelial cell (CFU-EC) and quantified at admission, at 24 and 72 hr, at day 7, and at 3 months. At the same time, serum levels of VEGF, SDF-1α, and active MMP-9 were measured by ELISA. The primary endpoint was EPC increment during the first week, which was defined as the difference in the number of CFU-EC between day 7 and admission. We found that VEGF (r = 0.782), SDF-1α (r = 0.828), and active MMP-9 (r = 0.740) levels at 24 hr from stroke onset showed a strong correlation with EPC increment. Similar results were found for VEGF levels at 72 hr (r = 0.839) and at day 7 (r = 0.602) as well as for active MMP-9 levels at 72 hr (r = 0.442) and at day 7 (r = 0.474). In the multivariate analyses, serum levels of VEGF at 72 hr (B: 0.074, P < 0.0001) and SDF-1α at 24 hr (B: 0.049, P = 0.008) were independent factors for EPC increment during the first week of evolution. These findings suggest that VEGF and SDF-1α may mediate EPC proliferation in human ischemic stroke.
Collapse
Affiliation(s)
- Tomás Sobrino
- Department of Neurology, Clinical Neurosciences Research Laboratory, Hospital Clínico Universitario, IDIS, University of Santiago de Compostela, Santiago de Compostela, Spain
| | | | | | | | | | | |
Collapse
|