1
|
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
|
2
|
Salaudeen MA, Allan S, Pinteaux E. Hypoxia and interleukin-1-primed mesenchymal stem/stromal cells as novel therapy for stroke. Hum Cell 2024; 37:154-166. [PMID: 37987924 PMCID: PMC10764391 DOI: 10.1007/s13577-023-00997-1] [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: 08/15/2023] [Accepted: 10/11/2023] [Indexed: 11/22/2023]
Abstract
Promising preclinical stroke research has not yielded meaningful and significant success in clinical trials. This lack of success has prompted the need for refinement of preclinical studies with the intent to optimize the chances of clinical success. Regenerative medicine, especially using mesenchymal stem/stromal cells (MSCs), has gained popularity in the last decade for treating many disorders, including central nervous system (CNS), such as stroke. In addition to less stringent ethical constraints, the ample availability of MSCs also makes them an attractive alternative to totipotent and other pluripotent stem cells. The ability of MSCs to differentiate into neurons and other brain parenchymal and immune cells makes them a promising therapy for stroke. However, these cells also have some drawbacks that, if not addressed, will render MSCs unfit for treating ischaemic stroke. In this review, we highlighted the molecular and cellular changes that occur following an ischaemic stroke (IS) incidence and discussed the physiological properties of MSCs suitable for tackling these changes. We also went further to discuss the major drawbacks of utilizing MSCs in IS and how adequate priming using both hypoxia and interleukin-1 can optimize the beneficial properties of MSCs while eliminating these drawbacks.
Collapse
Affiliation(s)
- Maryam Adenike Salaudeen
- Faculty of Biology, Medicine, and Health, Division of Neuroscience, University of Manchester, Manchester, UK
- Department of Pharmacology and Therapeutics, Ahmadu Bello University, Zaria, Nigeria
| | - Stuart Allan
- Faculty of Biology, Medicine, and Health, Division of Neuroscience, University of Manchester, Manchester, UK
| | - Emmanuel Pinteaux
- Faculty of Biology, Medicine, and Health, Division of Neuroscience, University of Manchester, Manchester, UK.
| |
Collapse
|
3
|
Zhou L, Wang J, Huang J, Song X, Wu Y, Chen X, Tan Y, Yang Q. The role of mesenchymal stem cell transplantation for ischemic stroke and recent research developments. Front Neurol 2022; 13:1000777. [PMID: 36468067 PMCID: PMC9708730 DOI: 10.3389/fneur.2022.1000777] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Accepted: 10/03/2022] [Indexed: 09/08/2023] Open
Abstract
Ischemic stroke is a common cerebrovascular disease that seriously affects human health. However, most patients do not practice self-care and cannot rely on the current clinical treatment for guaranteed functional recovery. Stem cell transplantation is an emerging treatment studied in various central nervous system diseases. More importantly, animal studies show that transplantation of mesenchymal stem cells (MSCs) can alleviate neurological deficits and bring hope to patients suffering from ischemic stroke. This paper reviews the biological characteristics of MSCs and discusses the mechanism and progression of MSC transplantation to provide new therapeutic directions for ischemic stroke.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Qin Yang
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
4
|
Patkar S, Uwanogho D, Modo M, Tate RJ, Plevin R, Carswell HVO. Targeting 17β-estradiol biosynthesis in neural stem cells improves stroke outcome. Front Cell Neurosci 2022; 16:917181. [PMID: 35936502 PMCID: PMC9355602 DOI: 10.3389/fncel.2022.917181] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2022] [Accepted: 06/27/2022] [Indexed: 11/18/2022] Open
Abstract
Dax-1 (dosage-sensitive sex reversal adrenal hypoplasia congenital region on X-chromosome gene 1) blocks 17β-estradiol biosynthesis and its knockdown would be expected to increase 17β-estradiol production. We hypothesized that knockdown of Dax-1 in a conditionally immortalized neural stem cell (NSC) line, MHP36, is a useful approach to increase 17β-estradiol production. Short hairpin (sh) RNA targeted to Dax-1 in NSCs, namely MHP36-Dax1KD cells, resulted in the degradation of Dax-1 RNA and attenuation of Dax-1 protein expression. In vitro, MHP36-Dax1KD cells exhibited overexpression of aromatase and increased 17β-estradiol secretion compared to MHP36 cells. As 17β-estradiol has been shown to promote the efficacy of cell therapy, we interrogated the application of 17β-estradiol-enriched NSCs in a relevant in vivo disease model. We hypothesized that MHP36-Dax1KD cells will enhance functional recovery after transplantation in a stroke model. C57BL/6 male adult mice underwent ischemia/reperfusion by left middle cerebral artery occlusion for 45 min using an intraluminal thread. Two days later male mice randomly received vehicle, MHP36 cells, MHP36-Dax1KD cells, and MHP36 cells suspended in 17β-estradiol (100 nm) or 17β-estradiol alone (100 nm) with serial behavioral testing over 28 days followed by post-mortem histology and blinded analysis. Recovery of sensorimotor function was accelerated and enhanced, and lesion volume was reduced by MHP36-Dax1KD transplants. Regarding mechanisms, immunofluorescence indicated increased synaptic plasticity and neuronal differentiation after MHP36-Dax1KD transplants. In conclusion, knockdown of Dax-1 is a useful target to increase 17β-estradiol biosynthesis in NSCs and improves functional recovery after stroke in vivo, possibly mediated through neuroprotection and improved synaptic plasticity. Therefore, targeting 17β-estradiol biosynthesis in stem cells may be a promising therapeutic strategy for enhancing the efficacy of stem cell-based therapies for stroke.
Collapse
Affiliation(s)
- Shalmali Patkar
- Strathclyde Institute of Pharmacy and Biological Sciences, University of Strathclyde, Glasgow, United Kingdom
| | - Dafe Uwanogho
- Department of Neuroscience, James Black Centre, King’s College London, London, United Kingdom
| | - Michel Modo
- Department of Neuroscience, James Black Centre, King’s College London, London, United Kingdom
- Department of Radiology, University of Pittsburgh, Pittsburgh, PA, United States
| | - Rothwelle J. Tate
- Strathclyde Institute of Pharmacy and Biological Sciences, University of Strathclyde, Glasgow, United Kingdom
| | - Robin Plevin
- Strathclyde Institute of Pharmacy and Biological Sciences, University of Strathclyde, Glasgow, United Kingdom
| | - Hilary V. O. Carswell
- Strathclyde Institute of Pharmacy and Biological Sciences, University of Strathclyde, Glasgow, United Kingdom
- *Correspondence: Hilary V. O. Carswell
| |
Collapse
|
5
|
Ischemic Brain Stroke and Mesenchymal Stem Cells: An Overview of Molecular Mechanisms and Therapeutic Potential. Stem Cells Int 2022; 2022:5930244. [PMID: 35663353 PMCID: PMC9159823 DOI: 10.1155/2022/5930244] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Revised: 10/12/2021] [Accepted: 05/04/2022] [Indexed: 12/15/2022] Open
Abstract
Ischemic brain injury is associated with a high rate of mortality and disability with no effective therapeutic strategy. Recently, a growing number of studies are focusing on mesenchymal stem cell-based therapies for neurodegenerative disorders. However, despite having the promising outcome of preclinical studies, the clinical application of stem cell therapy remained elusive due to little or no progress in clinical trials. The objective of this study was to provide a generalized critique for the role of mesenchymal stem cell therapy in ischemic stroke injury, its underlying mechanisms, and constraints on its preclinical and clinical applications. Thus, we attempted to present an overview of previously published reports to evaluate the progress and provide molecular basis of mesenchymal stem cells (MSCs) therapy and its application in preclinical and clinical settings, which could aid in designing an effective regenerative therapeutic strategy in the future.
Collapse
|
6
|
Liang CC, Shaw SW, Huang YH, Lee TH. Human amniotic fluid stem cells can improve cerebral vascular remodelling and neurological function after focal cerebral ischaemia in diabetic rats. J Cell Mol Med 2021; 25:10185-10196. [PMID: 34622573 PMCID: PMC8572791 DOI: 10.1111/jcmm.16956] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 09/12/2021] [Accepted: 09/23/2021] [Indexed: 01/04/2023] Open
Abstract
Diabetes causes vascular injury and carries a high risk of ischaemic stroke. Human amniotic fluid stem cells (hAFSCs) can enhance cerebral vascular remodelling and have the potential to improve neurological function after stroke in diabetic rats. Five groups of female rats were examined: (1) normal control, (2) type 1 diabetic (T1DM) rats induced by streptozotocin injection (DM), (3) non‐DM rats receiving 60‐minute middle cerebral artery occlusion (MCAO), (4) T1DM rats receiving 60‐minute MCAO (DM + MCAO) and (5) T1DM rats receiving 60‐minute MCAO and injection with 5 × 106 hAFSCs at 3 h after MCAO (DM + MCAO + hAFSCs). Neurological function was examined before, and at 1, 7, 14, 21 and 28 days, and cerebral infarction volume and haemorrhage, cerebral vascular density, angiogenesis and inflammatory were examined at 7 and 28 days after MCAO. hAFSCs treatment caused a significant improvement of neurological dysfunction, infarction volume, blood‐brain barrier leakage, cerebral arterial density, vascular density and angiogenesis and a reduction of brain haemorrhage and inflammation compared with non‐treatment. Our results showed that the effect of hAFSCs treatment against focal cerebral ischaemia may act through the recovery of vascular remodelling and angiogenesis and the reduction of inflammation in ischaemic brain.
Collapse
Affiliation(s)
- Ching-Chung Liang
- Urogynecology Section, Department of Obstetrics and Gynecology, Chang Gung Memorial Hospital, Linkou Medical Center, Taoyuan, Taiwan.,College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Steven W Shaw
- College of Medicine, Chang Gung University, Taoyuan, Taiwan.,Division of Obstetrics, Department of Obstetrics and Gynecology, Taipei Chang Gung Memorial Hospital, Taipei, Taiwan.,Prenatal Cell and Gene Therapy Group, Institute for Women's Health, University College London, London, UK
| | - Yung-Hsin Huang
- Urogynecology Section, Department of Obstetrics and Gynecology, Chang Gung Memorial Hospital, Linkou Medical Center, Taoyuan, Taiwan
| | - Tsong-Hai Lee
- College of Medicine, Chang Gung University, Taoyuan, Taiwan.,Stroke Center and Department of Neurology, Chang Gung Memorial Hospital, Linkou Medical Center, Taoyuan, Taiwan
| |
Collapse
|
7
|
Zhang S, Lachance BB, Moiz B, Jia X. Optimizing Stem Cell Therapy after Ischemic Brain Injury. J Stroke 2020; 22:286-305. [PMID: 33053945 PMCID: PMC7568970 DOI: 10.5853/jos.2019.03048] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Accepted: 06/17/2020] [Indexed: 12/13/2022] Open
Abstract
Stem cells have been used for regenerative and therapeutic purposes in a variety of diseases. In ischemic brain injury, preclinical studies have been promising, but have failed to translate results to clinical trials. We aimed to explore the application of stem cells after ischemic brain injury by focusing on topics such as delivery routes, regeneration efficacy, adverse effects, and in vivo potential optimization. PUBMED and Web of Science were searched for the latest studies examining stem cell therapy applications in ischemic brain injury, particularly after stroke or cardiac arrest, with a focus on studies addressing delivery optimization, stem cell type comparison, or translational aspects. Other studies providing further understanding or potential contributions to ischemic brain injury treatment were also included. Multiple stem cell types have been investigated in ischemic brain injury treatment, with a strong literature base in the treatment of stroke. Studies have suggested that stem cell administration after ischemic brain injury exerts paracrine effects via growth factor release, blood-brain barrier integrity protection, and allows for exosome release for ischemic injury mitigation. To date, limited studies have investigated these therapeutic mechanisms in the setting of cardiac arrest or therapeutic hypothermia. Several delivery modalities are available, each with limitations regarding invasiveness and safety outcomes. Intranasal delivery presents a potentially improved mechanism, and hypoxic conditioning offers a potential stem cell therapy optimization strategy for ischemic brain injury. The use of stem cells to treat ischemic brain injury in clinical trials is in its early phase; however, increasing preclinical evidence suggests that stem cells can contribute to the down-regulation of inflammatory phenotypes and regeneration following injury. The safety and the tolerability profile of stem cells have been confirmed, and their potent therapeutic effects make them powerful therapeutic agents for ischemic brain injury patients.
Collapse
Affiliation(s)
- Shuai Zhang
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Brittany Bolduc Lachance
- Program in Trauma, Department of Neurology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Bilal Moiz
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Xiaofeng Jia
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD, USA.,Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD, USA.,Department of Orthopedics, University of Maryland School of Medicine, Baltimore, MD, USA.,Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
8
|
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: 171] [Impact Index Per Article: 24.4] [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
|
9
|
Liang CC, Shaw SWS, Huang YH, Lin YH, Lee TH. Bladder Transplantation of Amniotic Fluid Stem Cell may Ameliorate Bladder Dysfunction After Focal Cerebral Ischemia in Rat. Stem Cells Transl Med 2017; 6:1227-1236. [PMID: 28186672 PMCID: PMC5442832 DOI: 10.1002/sctm.16-0212] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2016] [Revised: 07/29/2016] [Accepted: 10/31/2016] [Indexed: 12/20/2022] Open
Abstract
The objective is to investigate whether human amniotic fluid stem cells (hAFSCs) grafting into the bladder may influence bladder functional and molecular changes in an animal stroke model. Female rats were divided into three groups: sham, middle cerebral artery occlusion (MCAO) alone, and MCAO plus 1 × 106 hAFSCs transplanting into bladder wall. Bladder function was analyzed by cystometry at days 3 and 10 after MCAO. The expressions of bladder nerve growth factor (NGF), M2‐muscarinic, M3‐muscarinic, and P2X1 receptors were measured by immunohistochemistry and real‐time polymerase chain reaction. When compared with sham‐operated group, MCAO alone rats had significant increase in residual volume and decrease in voided volume and intercontraction interval; however, these bladder dysfunctions were improved following hAFSCs transplantation. The immunoreactivities of NGF, M3, and P2X1 significantly decreased at days 3 and 10, but M2 increased at day 3 after MCAO. Following hAFSCs transplantation, the immunoreactivities of NGF and P2X1 significantly increased at day 3, and M2 increased at day 10 after MCAO. The mRNAs of NGF, M2, and M3 significantly increased at day 3, but NGF and M2 decreased at day 10 after MCAO. Following hAFSCs transplantation, there was significant decrease in M2 mRNA at day 3 and increase in P2X1 mRNA at days 3 and 10 after MCAO. Bladder dysfunction caused by MCAO can be improved by hAFSCs transplanting into bladder which may be related to the expressions of bladder NGF, and muscarinic and P2X1 receptors. Stem Cells Translational Medicine2017;6:1227–1236
Collapse
Affiliation(s)
- Ching-Chung Liang
- Female Urology Section, Department of Obstetrics and Gynecology, Chang Gung Memorial Hospital Linkou Medical Center, Taoyuan, Taiwan, Republic of China.,College of Medicine, Chang Gung University, Taoyuan, Taiwan, Republic of China
| | - S W Steven Shaw
- Division of Obstetrics, Department of Obstetrics and Gynecology, Chang Gung Memorial Hospital Linkou Medical Center, Taoyuan, Taiwan, Republic of China.,College of Medicine, Chang Gung University, Taoyuan, Taiwan, Republic of China.,Prenatal Cell and Gene Therapy Group, Institute for Women's Health, University College London, London, United Kingdom
| | - Yung-Hsin Huang
- Female Urology Section, Department of Obstetrics and Gynecology, Chang Gung Memorial Hospital Linkou Medical Center, Taoyuan, Taiwan, Republic of China
| | - Yi-Hao Lin
- Female Urology Section, Department of Obstetrics and Gynecology, Chang Gung Memorial Hospital Linkou Medical Center, Taoyuan, Taiwan, Republic of China.,College of Medicine, Chang Gung University, Taoyuan, Taiwan, Republic of China
| | - Tsong-Hai Lee
- Stroke Center and Department of Neurology, Chang Gung Memorial Hospital Linkou Medical Center, Taoyuan, Taiwan, Republic of China.,College of Medicine, Chang Gung University, Taoyuan, Taiwan, Republic of China
| |
Collapse
|
10
|
Hassanein SMA, Nasr Eldin MH, Amer HA, Abdelhamid AE, El Houssinie M, Ibrahim A. Human Umbilical Cord Blood CD34-Positive Cells as Predictors of the Incidence and Short-Term Outcome of Neonatal Hypoxic-Ischemic Encephalopathy: A Pilot Study. J Clin Neurol 2017; 13:84-90. [PMID: 28079317 PMCID: PMC5242164 DOI: 10.3988/jcn.2017.13.1.84] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2016] [Revised: 11/04/2016] [Accepted: 11/07/2016] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND AND PURPOSE Neonatal hypoxic-ischemic encephalopathy (HIE) is one of the leading causes of neurological handicap in developing countries. Human umbilical cord blood (hUCB) CD34-positive (CD34⁺) stem cells exhibit the potential for neural repair. We tested the hypothesis that hUCB CD34⁺ stem cells and other cell types [leukocytes and nucleated red blood cells (NRBCs)] that are up-regulated during the acute stage of perinatal asphyxia (PA) could play a role in the early prediction of the occurrence, severity, and mortality of HIE. METHODS This case-control pilot study investigated consecutive neonates exposed to PA. The hUCB CD34⁺ cell count in mononuclear layers was assayed using a flow cytometer. Twenty full-term neonates with PA and 25 healthy neonates were enrolled in the study. RESULTS The absolute CD34⁺ cell count (p=0.02) and the relative CD34⁺ cell count (CD34⁺%) (p<0.001) in hUCB were higher in the HIE patients (n=20) than the healthy controls. The hUCB absolute CD34⁺ cell count (p=0.04), CD34⁺% (p<0.01), and Hobel risk scores (p=0.04) were higher in patients with moderate-to-severe HIE (n=9) than in those with mild HIE (n=11). The absolute CD34⁺ cell count was strongly correlated with CD34⁺% (p<0.001), Hobel risk score (p=0.04), total leukocyte count (TLC) (p<0.001), and NRBC count (p=0.01). CD34⁺% was correlated with TLC (p=0.02). CONCLUSIONS hUCB CD34⁺ cells can be used to predict the occurrence, severity, and mortality of neonatal HIE after PA.
Collapse
Affiliation(s)
- Sahar M A Hassanein
- Department of Pediatrics, Faculty of Medicine, Ain Shams University, Cairo, Egypt.
| | | | - Hanaa A Amer
- Department of Clinical Pathology, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | | | - Moustafa El Houssinie
- Department of Community Medicine, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | | |
Collapse
|