1
|
Yoshida Y, Takeda Y, Yamahara K, Yamamoto H, Takagi T, Kuramoto Y, Nakano-Doi A, Nakagomi T, Soma T, Matsuyama T, Doe N, Yoshimura S. Enhanced angiogenic properties of umbilical cord blood primed by OP9 stromal cells ameliorates neurological deficits in cerebral infarction mouse model. Sci Rep 2023; 13:262. [PMID: 36609640 PMCID: PMC9822952 DOI: 10.1038/s41598-023-27424-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Accepted: 12/29/2022] [Indexed: 01/09/2023] Open
Abstract
Umbilical cord blood (UCB) transplantation shows proangiogenic effects and contributes to symptom amelioration in animal models of cerebral infarction. However, the effect of specific cell types within a heterogeneous UCB population are still controversial. OP9 is a stromal cell line used as feeder cells to promote the hematoendothelial differentiation of embryonic stem cells. Hence, we investigated the changes in angiogenic properties, underlying mechanisms, and impact on behavioral deficiencies caused by cerebral infarction in UCB co-cultured with OP9 for up to 24 h. In the network formation assay, only OP9 pre-conditioned UCB formed network structures. Single-cell RNA sequencing and flow cytometry analysis showed a prominent phenotypic shift toward M2 in the monocytic fraction of OP9 pre-conditioned UCB. Further, OP9 pre-conditioned UCB transplantation in mice models of cerebral infarction facilitated angiogenesis in the peri-infarct lesions and ameliorated the associated symptoms. In this study, we developed a strong, fast, and feasible method to augment the M2, tissue-protecting, pro-angiogenic features of UCB using OP9. The ameliorative effect of OP9-pre-conditioned UCB in vivo could be partly due to promotion of innate angiogenesis in peri-infarct lesions.
Collapse
Affiliation(s)
- Yasunori Yoshida
- grid.272264.70000 0000 9142 153XDepartment of Neurosurgery, Hyogo Medical University, 1-1 Mukogawa, Nishinomiya, Hyogo 663-8501 Japan
| | - Yuki Takeda
- grid.272264.70000 0000 9142 153XDepartment of Neurosurgery, Hyogo Medical University, 1-1 Mukogawa, Nishinomiya, Hyogo 663-8501 Japan
| | - Kenichi Yamahara
- Laboratory of Molecular and Cellular Therapy, Institute for Advanced Medical Sciences, Hyogo Medical University, 1-1 Mukogawa, Nishinomiya, Hyogo, 663-8501, Japan.
| | - Hanae Yamamoto
- grid.272264.70000 0000 9142 153XLaboratory of Molecular and Cellular Therapy, Institute for Advanced Medical Sciences, Hyogo Medical University, 1-1 Mukogawa, Nishinomiya, Hyogo 663-8501 Japan
| | - Toshinori Takagi
- grid.272264.70000 0000 9142 153XDepartment of Neurosurgery, Hyogo Medical University, 1-1 Mukogawa, Nishinomiya, Hyogo 663-8501 Japan
| | - Yoji Kuramoto
- grid.272264.70000 0000 9142 153XDepartment of Neurosurgery, Hyogo Medical University, 1-1 Mukogawa, Nishinomiya, Hyogo 663-8501 Japan
| | - Akiko Nakano-Doi
- Laboratory of Neurogenesis and CNS Repair, Institute for Advanced Medical Sciences, Hyogo Medial University, 1-1 Mukogawa, Nishinomiya, Hyogo 663-8501 Japan
| | - Takayuki Nakagomi
- Laboratory of Neurogenesis and CNS Repair, Institute for Advanced Medical Sciences, Hyogo Medial University, 1-1 Mukogawa, Nishinomiya, Hyogo 663-8501 Japan
| | - Toshihiro Soma
- grid.272264.70000 0000 9142 153XDepartment of Hematology, Hyogo Medical University, 1-1 Mukogawa, Nishinomiya, Hyogo 663-8501 Japan
| | - Tomohiro Matsuyama
- grid.272264.70000 0000 9142 153XDepartment of Therapeutic Progress in Brain Diseases, Hyogo Medical University, 1-1 Mukogawa, Nishinomiya, Hyogo 663-8501 Japan
| | - Nobutaka Doe
- Laboratory of Neurogenesis and CNS Repair, Institute for Advanced Medical Sciences, Hyogo Medial University, 1-1 Mukogawa, Nishinomiya, Hyogo 663-8501 Japan ,grid.272264.70000 0000 9142 153XDepartment of Occupational Therapy, School of Rehabilitation, Hyogo Medical University, 1-3-6 Minatojima, Chuo-Ku, Kobe, Hyogo 650-8530 Japan
| | - Shinichi Yoshimura
- grid.272264.70000 0000 9142 153XDepartment of Neurosurgery, Hyogo Medical University, 1-1 Mukogawa, Nishinomiya, Hyogo 663-8501 Japan
| |
Collapse
|
2
|
Salehi MS, Safari A, Pandamooz S, Jurek B, Hooshmandi E, Owjfard M, Bayat M, Zafarmand SS, Miyan JA, Borhani-Haghighi A. The Beneficial Potential of Genetically Modified Stem Cells in the Treatment of Stroke: a Review. Stem Cell Rev Rep 2022; 18:412-440. [PMID: 34033001 PMCID: PMC8144279 DOI: 10.1007/s12015-021-10175-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/26/2021] [Indexed: 12/16/2022]
Abstract
The last two decades have witnessed a surge in investigations proposing stem cells as a promising strategy to treat stroke. Since growth factor release is considered as one of the most important aspects of cell-based therapy, stem cells over-expressing growth factors are hypothesized to yield higher levels of therapeutic efficiency. In pre-clinical studies of the last 15 years that were investigating the efficiency of stem cell therapy for stroke, a variety of stem cell types were genetically modified to over-express various factors. In this review we summarize the current knowledge on the therapeutic efficiency of stem cell-derived growth factors, encompassing techniques employed and time points to evaluate. In addition, we discuss several types of stem cells, including the recently developed model of epidermal neural crest stem cells, and genetically modified stem cells over-expressing specific factors, which could elevate the restorative potential of naive stem cells. The restorative potential is based on enhanced survival/differentiation potential of transplanted cells, apoptosis inhibition, infarct volume reduction, neovascularization or functional improvement. Since the majority of studies have focused on the short-term curative effects of genetically engineered stem cells, we emphasize the need to address their long-term impact.
Collapse
Affiliation(s)
- Mohammad Saied Salehi
- Clinical Neurology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Anahid Safari
- Stem Cells Technology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Sareh Pandamooz
- Stem Cells Technology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Benjamin Jurek
- Institute of Molecular and Cellular Anatomy, University of Regensburg, Regensburg, Germany
| | - Etrat Hooshmandi
- Clinical Neurology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Maryam Owjfard
- Clinical Neurology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mahnaz Bayat
- Clinical Neurology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | | | - Jaleel A Miyan
- Faculty of Biology, Division of Neuroscience & Experimental Psychology, The University of Manchester, Manchester, UK
| | | |
Collapse
|
3
|
Mazen NF, Abdel‐Fattah EA, Desoky SR, El‐Shal AS. Therapeutic role of adipose tissue-derived stem cells versus microvesicles in a rat model of cerebellar injury. J Cell Mol Med 2022; 26:326-342. [PMID: 34874117 PMCID: PMC8743657 DOI: 10.1111/jcmm.17083] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Revised: 11/10/2021] [Accepted: 11/17/2021] [Indexed: 12/12/2022] Open
Abstract
Monosodium glutamate (MSG) is a controversial food additive reported to cause negative effects on public health. Adipose stem cells (ASCs) and their derived vesicles (MVs) represent a promising cure for human diseases. This work was planned to compare the therapeutic effects of adipose stem cells and microvesicles in MSG-induced cerebellar damage. Forty adult healthy male Wister rats were equally divided into four groups: Group I (control group), group II (MSG-treated), group III (MSG/ASCs-treated), and group IV (MSG/MVs-treated). Motor behaviour of rats was assessed. Characterization of ASCs and MVs was done by flow cytometry. The cerebellum was processed for light and electron microscopic studies, and immunohistochemical localization of PCNA and GFAP. Morphometry was done for the number of Purkinje cells in H&E-stained sections, area per cent of GFAP immune reactivity and number of positive PCNA cells. Our results showed MSG-induced deterioration in the motor part. Moreover, MSG increases oxidant and apoptotic with decreases of antioxidant biomarkers. Structural changes in the cerebellar cortex as degeneration of nerve cells and gliosis were detected. There were also a decrease in the number of Purkinje cells, an increase in the area per cent of GFAP immune reactivity and a decrease in the number of positive PCNA cells, as compared to the control. Rats treated with ASCs showed marked functional and structural improvement in comparison with MV-treated rats. Thus, both ASCs and MVs had therapeutic potential for MSG-induced cerebellar damage with better results in case of ASCs.
Collapse
Affiliation(s)
- Nehad F. Mazen
- Medical Histology and Cell Biology DepartmentFaculty of MedicineZagazig UniversityZagazigEgypt
| | - Eman A. Abdel‐Fattah
- Medical Histology and Cell Biology DepartmentFaculty of MedicineZagazig UniversityZagazigEgypt
| | - Shimaa R. Desoky
- Histology and Cell Biology DepartmentFaculty of MedicineSuez UniversityIsmailiaEgypt
| | - Amal S. El‐Shal
- Medical Biochemistry & Molecular Biology DepartmentFaculty of Human MedicineZagazig UniversityZagazigEgypt
| |
Collapse
|
4
|
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: 41] [Impact Index Per Article: 8.2] [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
|
5
|
Steliga A, Kowiański P, Czuba E, Waśkow M, Moryś J, Lietzau G. Neurovascular Unit as a Source of Ischemic Stroke Biomarkers-Limitations of Experimental Studies and Perspectives for Clinical Application. Transl Stroke Res 2020; 11:553-579. [PMID: 31701356 PMCID: PMC7340668 DOI: 10.1007/s12975-019-00744-5] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Revised: 09/23/2019] [Accepted: 09/24/2019] [Indexed: 01/13/2023]
Abstract
Cerebral stroke, which is one of the most frequent causes of mortality and leading cause of disability in developed countries, often leads to devastating and irreversible brain damage. Neurological and neuroradiological diagnosis of stroke, especially in its acute phase, is frequently uncertain or inconclusive. This results in difficulties in identification of patients with poor prognosis or being at high risk for complications. It also makes difficult identification of these stroke patients who could benefit from more aggressive therapies. In contrary to the cardiovascular disease, no single biomarker is available for the ischemic stroke, addressing the abovementioned issues. This justifies the need for identifying of effective diagnostic measures characterized by high specificity and sensitivity. One of the promising avenues in this area is studies on the panels of biomarkers characteristic for processes which occur in different types and phases of ischemic stroke and represent all morphological constituents of the brains' neurovascular unit (NVU). In this review, we present the current state of knowledge concerning already-used or potentially applicable biomarkers of the ischemic stroke. We also discuss the perspectives for identification of biomarkers representative for different types and phases of the ischemic stroke, as well as for different constituents of NVU, which concentration levels correlate with extent of brain damage and patients' neurological status. Finally, a critical analysis of perspectives on further improvement of the ischemic stroke diagnosis is presented.
Collapse
Affiliation(s)
- Aleksandra Steliga
- Faculty of Health Sciences, Pomeranian University of Slupsk, 64 Bohaterów Westerplatte St., 76-200, Slupsk, Poland
| | - Przemysław Kowiański
- Faculty of Health Sciences, Pomeranian University of Slupsk, 64 Bohaterów Westerplatte St., 76-200, Slupsk, Poland.
- Department of Anatomy and Neurobiology, Medical University of Gdansk, 1 Debinki St., 80-211, Gdansk, Poland.
| | - Ewelina Czuba
- Department of Anatomy and Neurobiology, Medical University of Gdansk, 1 Debinki St., 80-211, Gdansk, Poland
| | - Monika Waśkow
- Faculty of Health Sciences, Pomeranian University of Slupsk, 64 Bohaterów Westerplatte St., 76-200, Slupsk, Poland
| | - Janusz Moryś
- Department of Anatomy and Neurobiology, Medical University of Gdansk, 1 Debinki St., 80-211, Gdansk, Poland
| | - Grażyna Lietzau
- Department of Anatomy and Neurobiology, Medical University of Gdansk, 1 Debinki St., 80-211, Gdansk, Poland
- Department of Clinical Science and Education, Södersjukhuset, Internal Medicine, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
6
|
Pischiutta F, Sammali E, Parolini O, Carswell HVO, Zanier ER. Placenta-Derived Cells for Acute Brain Injury. Cell Transplant 2019; 27:151-167. [PMID: 29562781 PMCID: PMC6434489 DOI: 10.1177/0963689717732992] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Acute brain injury resulting from ischemic/hemorrhagic or traumatic damage is one of the leading causes of mortality and disability worldwide and is a significant burden to society. Neuroprotective options to counteract brain damage are very limited in stroke and traumatic brain injury (TBI). Given the multifaceted nature of acute brain injury and damage progression, several therapeutic targets may need to be addressed simultaneously to interfere with the evolution of the injury and improve the patient’s outcome. Stem cells are ideal candidates since they act on various mechanisms of protection and repair, improving structural and functional outcomes after experimental stroke or TBI. Stem cells isolated from placenta offer advantages due to their early embryonic origin, ease of procurement, and ethical acceptance. We analyzed the evidence for the beneficial effects of placenta-derived stem cells in acute brain injury, with the focus on experimental studies of TBI and stroke, the engineering strategies pursued to foster cell potential, and characterization of the bioactive molecules secreted by placental cells, known as their secretome, as an alternative cell-free strategy. Results from the clinical application of placenta-derived stem cells for acute brain injury and ongoing clinical trials are summarily discussed.
Collapse
Affiliation(s)
- Francesca Pischiutta
- 1 Department of Neuroscience, Laboratory of Acute Brain Injury and Therapeutic Strategies, IRCCS Istituto di Ricerche Farmacologiche Mario Negri, Milan, Italy
| | - Eliana Sammali
- 1 Department of Neuroscience, Laboratory of Acute Brain Injury and Therapeutic Strategies, IRCCS Istituto di Ricerche Farmacologiche Mario Negri, Milan, Italy.,2 Department of Cerebrovascular Diseases, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Ornella Parolini
- 3 Centro di Ricerca E. Menni, Fondazione Poliambulanza Istituto Ospedaliero, Brescia, Italy.,4 Institute of Anatomy and Cell Biology, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Hilary V O Carswell
- 5 Strathclyde Institute of Pharmacy and Biomedical Sciences (SIPBS), University of Strathclyde, Glasgow, United Kingdom
| | - Elisa R Zanier
- 1 Department of Neuroscience, Laboratory of Acute Brain Injury and Therapeutic Strategies, IRCCS Istituto di Ricerche Farmacologiche Mario Negri, Milan, Italy
| |
Collapse
|
7
|
Wu KJ, Yu S, Lee JY, Hoffer B, Wang Y. Improving Neurorepair in Stroke Brain Through Endogenous Neurogenesis-Enhancing Drugs. Cell Transplant 2018; 26:1596-1600. [PMID: 29113469 PMCID: PMC5680955 DOI: 10.1177/0963689717721230] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Stroke induces not only cell death but also neurorepair. De novo neurogenesis has been found in the subventricular zone of the adult mammalian brain days after stroke. Most of these newly generated cells die shortly after the insult. Recent studies have shown that pharmacological manipulation can improve the survival of endogenous neuroprogenitor cells and neural regeneration in stroke rats. As these drugs target the endogenous reparative processes that occur days after stroke, they may provide a prolonged window for stroke therapy. Here, we discuss endogenous neurogenesis-enhancing drugs and review the general status of stroke therapeutics in evaluating the field of pharmacotherapy for stroke.
Collapse
Affiliation(s)
- Kuo-Jen Wu
- 1 Center for Neuropsychiatric Research, National Health Research Institutes, Miaoli, Taiwan
| | - Seongjin Yu
- 1 Center for Neuropsychiatric Research, National Health Research Institutes, Miaoli, Taiwan
| | - Jea-Young Lee
- 2 University of South Florida Morsani College of Medicine, FL, USA
| | - Barry Hoffer
- 3 Case Western Reserve University, Cleveland, OH, USA
| | - Yun Wang
- 1 Center for Neuropsychiatric Research, National Health Research Institutes, Miaoli, Taiwan
| |
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: 176] [Impact Index Per Article: 22.0] [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
|
Dewan SN, Wang Y, Yu S. Drug treatments that optimize endogenous neurogenesis as a therapeutic option for stroke. Brain Circ 2017; 3:152-155. [PMID: 30276317 PMCID: PMC6057687 DOI: 10.4103/bc.bc_20_17] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2017] [Revised: 09/03/2017] [Accepted: 09/05/2017] [Indexed: 01/28/2023] Open
Abstract
Cell death and neurogenesis have been examined after stroke in the subventricular zone of the adult mammalian brain. New research focuses on the use of drugs to improve the viability of neural progenitor cells in rats after stroke. The aim of the drugs is to lengthen the timeframe for stroke therapy by targeting the endogenous repair mechanism that follows injury. In this paper, we look at the broad state of stroke therapy to assess the effectiveness of endogenous neurogenesis-enhancing drugs on stroke. This paper is a review article. Referred literature in this paper has been listed in the reference section. The data sets supporting the conclusions of this article are available online by searching various databases, including PubMed. Some original points in this article come from the laboratory practice in our research center and the authors’ experiences.
Collapse
Affiliation(s)
- Shyam N Dewan
- Center of Excellence for Aging and Brain Repair, University of South Florida, Tampa, FL, USA
| | - Yun Wang
- Center for Neuropsychiatric Research, National Health Research Institutes, Taiwan
| | - Seongjin Yu
- Center for Neuropsychiatric Research, National Health Research Institutes, Taiwan
| |
Collapse
|
10
|
Doursout MF, Liang Y, Schiess MC, Padilla A, Poindexter BJ, Hickson-Bick DLM, Bick RJ. Are Temporal Differences in GDNF and NOS Isoform Induction Contributors to Neurodegeneration? A Fluorescence Microscopy-Based Study. Open Neurol J 2016; 10:67-76. [PMID: 27651844 PMCID: PMC5009294 DOI: 10.2174/1874205x01610010067] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2016] [Revised: 05/24/2016] [Accepted: 06/21/2016] [Indexed: 11/22/2022] Open
Abstract
BACKGROUND Specific factors in Parkinson's disease have become targets as to their protective and degenerative effects. We have demonstrated that cytokines and PD-CSF detrimentally affect microglia and astrocyte growth. While glial cell-derived neurotrophic factor (GDNF) has been recognized as a possible neuron-rescue agent, nitric oxide synthase (NOS) has been implicated in neurodegenerative processes. OBJECTIVE To demonstrate that glial cell activation, cytokine production, and NOS induction, play an intimate role in the loss of dopaminergic signaling, via mechanisms that are a result of inflammation and inflammatory stimuli. METHODS Study animals were sacrificed following endotoxin treatment and tissue sections were harvested and probed for GDNF and NOS isomers by fluorescence deconvolution microscopy. Fluorescence was mapped and quantified for each probe. RESULTS An immune cell influx into 'vulnerable' areas of the brain was seen, and three NOS isomers, inducible (iNOS), neuronal (nNOS) and endothelial (eNOS), were synthesized in the brains, a finding which suggests that each isomer has a role in neurodegeneration. eNOS was found associated with blood vessels, while iNOS was associated with glial and matrix cells and nNOS was located with both glia and neurons. Following endotoxin treatment, serum levels of nitric oxide were higher at 6-8 hours, while tissue levels of NOS were elevated for much longer. Thus, induction of NOS occurred earlier than the induction of GDNF. CONCLUSION Our findings suggest that the protective abilities of GDNF to combat neural destruction are not available rapidly enough, and do not remain at sufficiently high levels long enough to assert its protective effects. (250).
Collapse
Affiliation(s)
| | - Yangyan Liang
- Department of Anesthesiology, University of Texas McGovern Medical School, Houston,Texas, USA
| | - Mya C Schiess
- Department of Neurology, University of Texas McGovern Medical School, Houston,Texas, USA
| | - Angelica Padilla
- Department of Neurology, University of Texas McGovern Medical School, Houston,Texas, USA
| | - Brian J Poindexter
- Department of Pathology, University of Texas McGovern Medical School, Houston,Texas, USA
| | - Diane L M Hickson-Bick
- Department of Pathology, University of Texas McGovern Medical School, Houston,Texas, USA
| | - Roger J Bick
- Department of Pathology, University of Texas McGovern Medical School, Houston,Texas, USA
| |
Collapse
|
11
|
Islamov RR, Rizvanov AA, Fedotova VY, Izmailov AA, Safiullov ZZ, Garanina EE, Salafutdinov II, Sokolov ME, Mukhamedyarov MA, Palotás A. Tandem Delivery of Multiple Therapeutic Genes Using Umbilical Cord Blood Cells Improves Symptomatic Outcomes in ALS. Mol Neurobiol 2016; 54:4756-4763. [PMID: 27495938 DOI: 10.1007/s12035-016-0017-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2016] [Accepted: 07/28/2016] [Indexed: 12/16/2022]
Abstract
Current treatment options of chronic, progressive degenerative neuropsychiatric conditions offer only marginal efficacy, and there is no therapy which arrests or even reverses these diseases. Interest in genetic engineering and cell-based approaches have constantly been increasing, although most of them so far proved to be fruitless or at best provided very slight clinical benefit. In the light of the highly complex patho-mechanisms of these maladies, the failure of drugs aimed at targeting single molecules is not surprising. In order to improve their effectiveness, the role of a unique triple-combination gene therapy was investigated in this study. Intravenous injection of human umbilical cord blood mononuclear cell (hUCBMC) cotransduced with adenoviral vectors expressing vascular endothelial growth factor (VEGF), glial cell-derived neurotrophic factor (GDNF), and neural cell adhesion molecule (NCAM) resulted in prominent increase of life span and performance in behavioral tests in amyotrophic lateral sclerosis (ALS). Expression of the recombinant genes in hUCBMCs was confirmed as soon as 5 days after transduction by RT-PCR, and cells were detectable for as long as 1 month after grafting in lumbar spinal cord by immunofluorescent staining. Xenotransplantation of cells into mice blood without any immunosuppression demonstrated a high level of hUCBMCs homing and survivability in the central nervous system (CNS), most conspicuously in the spinal cord, but not in the spleen or liver. This study confirms an increased addressed homing and notable survivability of triple-transfected cells in lumbar spinal cord, yielding a remarkably enhanced therapeutic potential of hUCBMCs overexpressing neurotrophic factors.
Collapse
Affiliation(s)
- Rustem Robertovich Islamov
- Kazan State Medical University, Kazan, Russia. .,Department of Biology, Kazan State Medical University, ul. Butlerova 49, R-420012, Kazan, Russia.
| | | | | | | | | | | | | | | | | | - András Palotás
- Kazan Federal University, Kazan, Russia. .,Asklepios-Med (private medical practice and research center), Kossuth Lajos sgt. 23, Szeged, H-6722, Hungary.
| |
Collapse
|
12
|
Lawton C, Acosta S, Watson N, Gonzales-Portillo C, Diamandis T, Tajiri N, Kaneko Y, Sanberg PR, Borlongan CV. Enhancing endogenous stem cells in the newborn via delayed umbilical cord clamping. Neural Regen Res 2015; 10:1359-62. [PMID: 26604879 PMCID: PMC4625484 DOI: 10.4103/1673-5374.165218] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
There is currently no consensus among clinicians and scientists over the appropriate or optimal timing for umbilical cord clamping. However, many clinical studies have suggested that delayed cord clamping is associated with various neonatal benefits including increased blood volume, reduced need for blood transfusion, increased cerebral oxygenation in pre-term infants, and decreased frequency of iron deficiency anemia in term infants. Human umbilical cord blood contains significant amounts of stem and progenitor cells and is currently used in the treatment of several life-threatening diseases. We propose that delayed cord clamping be encouraged as it enhances blood flow from the placenta to the neonate, which is accompanied by an increase supply of valuable stem and progenitor cells, as well as may improve blood oxygenation and increase blood volume, altogether reducing the infant's susceptibility to both neonatal and age-related diseases.
Collapse
Affiliation(s)
- Christopher Lawton
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery and Brain Repair, University of South Florida College of Medicine, Tampa, FL, USA
| | - Sandra Acosta
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery and Brain Repair, University of South Florida College of Medicine, Tampa, FL, USA
| | - Nate Watson
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery and Brain Repair, University of South Florida College of Medicine, Tampa, FL, USA
| | - Chiara Gonzales-Portillo
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery and Brain Repair, University of South Florida College of Medicine, Tampa, FL, USA
| | - Theo Diamandis
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery and Brain Repair, University of South Florida College of Medicine, Tampa, FL, USA
| | - Naoki Tajiri
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery and Brain Repair, University of South Florida College of Medicine, Tampa, FL, USA
| | - Yuji Kaneko
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery and Brain Repair, University of South Florida College of Medicine, Tampa, FL, USA
| | - Paul R Sanberg
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery and Brain Repair, University of South Florida College of Medicine, Tampa, FL, USA
| | - Cesar V Borlongan
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery and Brain Repair, University of South Florida College of Medicine, Tampa, FL, USA
| |
Collapse
|
13
|
Abstract
Brain injury continues to be one of the leading causes of disability worldwide. Despite decades of research, there is currently no pharmacologically effective treatment for preventing neuronal loss and repairing the brain. As a result, novel therapeutic approaches, such as cell-based therapies, are being actively pursued to repair tissue damage and restore neurological function after injury. In this study, we examined the neuroprotective potential of amniotic fluid (AF) single cell clones, engineered to secrete glial cell derived neurotrophic factor (AF-GDNF), both in vitro and in a surgically induced model of brain injury. Our results show that pre-treatment with GDNF significantly increases cell survival in cultures of AF cells or cortical neurons exposed to hydrogen peroxide. Since improving the efficacy of cell transplantation depends on enhanced graft cell survival, we investigated whether AF-GDNF cells seeded on polyglycolic acid (PGA) scaffolds could enhance graft survival following implantation into the lesion cavity. Encouragingly, the AF-GDNF cells survived longer than control AF cells in serum-free conditions and continued to secrete GDNF both in vitro and following implantation into the injured motor cortex. AF-GDNF implantation in the acute period following injury was sufficient to activate the MAPK/ERK signaling pathway in host neural cells in the peri-lesion area, potentially boosting endogenous neuroprotective pathways. These results were complemented with promising trends in beam walk tasks in AF-GDNF/PGA animals during the 7 day timeframe. Further investigation is required to determine whether significant behavioural improvement can be achieved at a longer timeframe.
Collapse
|
14
|
Jin X, Wang F, Liu X, Liang B, Chen Z, He J, Zhang H, Zhang J. Negative Correlation of CD34+Cells with Blood-Brain Barrier Permeability following Traumatic Brain Injury in a Rat Model. Microcirculation 2014; 21:696-702. [PMID: 24894113 DOI: 10.1111/micc.12150] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2014] [Accepted: 05/29/2014] [Indexed: 12/16/2022]
Affiliation(s)
- Xuelong Jin
- Department of Physiology; Tianjin Medical University; Tianjin China
| | - Feifei Wang
- Department of Physiology; Tianjin Medical University; Tianjin China
| | - Xingju Liu
- Department of Physiology; Tianjin Medical University; Tianjin China
| | - Bin Liang
- Department of Physiology; Tianjin Medical University; Tianjin China
| | - Zequn Chen
- Department of Physiology; Tianjin Medical University; Tianjin China
| | - Junfeng He
- Liuyang Hospital of Traditional Chinese Medicine; the Affiliated Hospital to Hunan University of Chinese Medicine; Liuyang China
| | - Hong Zhang
- Department of Radiology; Tianjin Chest Hospital; Tianjin China
| | - Jianning Zhang
- Department of Neurosurgery; Tianjin Medical University General Hospital; Tianjin China
| |
Collapse
|
15
|
Tajiri N, Acosta S, Portillo-Gonzales GS, Aguirre D, Reyes S, Lozano D, Pabon M, Dela Peña I, Ji X, Yasuhara T, Date I, Solomita MA, Antonucci I, Stuppia L, Kaneko Y, Borlongan CV. Therapeutic outcomes of transplantation of amniotic fluid-derived stem cells in experimental ischemic stroke. Front Cell Neurosci 2014; 8:227. [PMID: 25165432 PMCID: PMC4131212 DOI: 10.3389/fncel.2014.00227] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2014] [Accepted: 07/23/2014] [Indexed: 01/29/2023] Open
Abstract
Accumulating preclinical evidence suggests the use of amnion as a source of stem cells for investigations of basic science concepts related to developmental cell biology, but also for stem cells’ therapeutic applications in treating human disorders. We previously reported isolation of viable rat amniotic fluid-derived stem (AFS) cells. Subsequently, we recently reported the therapeutic benefits of intravenous transplantation of AFS cells in a rodent model of ischemic stroke. Parallel lines of investigations have provided safety and efficacy of stem cell therapy for treating stroke and other neurological disorders. This review article highlights the need for investigations of mechanisms underlying AFS cells’ therapeutic benefits and discusses lab-to-clinic translational gating items in an effort to optimize the clinical application of the cell transplantation for stroke.
Collapse
Affiliation(s)
- Naoki Tajiri
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine Tampa, FL, USA
| | - Sandra Acosta
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine Tampa, FL, USA
| | - Gabriel S Portillo-Gonzales
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine Tampa, FL, USA
| | - Daniela Aguirre
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine Tampa, FL, USA
| | - Stephanny Reyes
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine Tampa, FL, USA
| | - Diego Lozano
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine Tampa, FL, USA
| | - Mibel Pabon
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine Tampa, FL, USA
| | - Ike Dela Peña
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine Tampa, FL, USA
| | - Xunming Ji
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University Beijing, China
| | - Takao Yasuhara
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences Okayama, Japan
| | - Isao Date
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences Okayama, Japan
| | - Marianna A Solomita
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine Tampa, FL, USA
| | - Ivana Antonucci
- Laboratory of Molecular Genetics, DISPUTer, School of Medicine and Health Sciences, "G. d 'Annunzio" University Chieti-Pescara, Italy
| | - Liborio Stuppia
- Laboratory of Molecular Genetics, DISPUTer, School of Medicine and Health Sciences, "G. d 'Annunzio" University Chieti-Pescara, Italy
| | - Yuji Kaneko
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine Tampa, FL, USA
| | - Cesar V Borlongan
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine Tampa, FL, USA
| |
Collapse
|
16
|
Effects of intravenous administration of umbilical cord blood CD34(+) cells in a mouse model of neonatal stroke. Neuroscience 2014; 263:148-58. [PMID: 24444827 DOI: 10.1016/j.neuroscience.2014.01.018] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2013] [Revised: 01/06/2014] [Accepted: 01/06/2014] [Indexed: 12/16/2022]
Abstract
Neonatal stroke occurs in approximately 1/4000 live births and results in life-long neurological impairments: e.g., cerebral palsy. Currently, there is no evidence-based specific treatment for neonates with stroke. Several studies have reported the benefits of umbilical cord blood (UCB) cell treatment in rodent models of neonatal brain injury. However, all of the studies examined the effects of administering either the UCB mononuclear cell fraction or UCB-derived mesenchymal stem cells in neonatal rat models. The objective of this study was to examine the effects of human UCB CD34(+) cells (hematopoietic stem cell/endothelial progenitor cells) in a mouse model of neonatal stroke, which we recently developed. On postnatal day 12, immunocompromized (SCID) mice underwent permanent occlusion of the left middle cerebral artery (MCAO). Forty-eight hours after MCAO, human UCB CD34(+) cells (1×10(5)cells) were injected intravenously into the mice. The area in which cerebral blood flow (CBF) was maintained was temporarily larger in the cell-treated group than in the phosphate-buffered saline (PBS)-treated group at 24h after treatment. With cell treatment, the percent loss of ipsilateral hemispheric volume was significantly ameliorated (21.5±1.9%) compared with the PBS group (25.6±5.1%) when assessed at 7weeks after MCAO. The cell-treated group did not exhibit significant differences from the PBS group in either rotarod (238±46s in the sham-surgery group, 175±49s in the PBS group, 203±54s in the cell-treated group) or open-field tests. The intravenous administration of human UCB CD34(+) cells modestly reduced histological ischemic brain damage after neonatal stroke in mice, with a transient augmentation of CBF in the peri-infarct area.
Collapse
|
17
|
Abo-Grisha N, Essawy S, Abo-Elmatty DM, Abdel-Hady Z. Effects of intravenous human umbilical cord blood CD34+ stem cell therapy versus levodopa in experimentally induced Parkinsonism in mice. Arch Med Sci 2013; 9:1138-51. [PMID: 24482663 PMCID: PMC3902714 DOI: 10.5114/aoms.2013.39237] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2012] [Revised: 07/13/2012] [Accepted: 08/23/2012] [Indexed: 11/17/2022] Open
Abstract
INTRODUCTION Parkinsonism is a neurodegenerative disease with impaired motor function. The current research was directed to investigate the effect of CD34+ stem cells versus levodopa in 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced Parkinsonism. MATERIAL AND METHODS Mice were divided into 4 groups; saline-injected, MPTP: received four MPTP injections (20 mg/kg, i.p.) at 2 h intervals, MPTP groups treated with levodopa/carbidopa (100/10 mg/kg/twice/day for 28 days) or single intravenous injection of 10(6) CD34+ stem cells/mouse at day 7 and allowed to survive until the end of week 5. RESULTS Levodopa and stem cells improved MPTP-induced motor deficits; they abolished the difference in stride length, decreased percentage of foot slip errors and increased ambulation, activity factor and mobility duration in parkinsonian mice (p < 0.05). Further, they significantly (p < 0.05) increased striatal dopamine (85.3 ±4.3 and 110.6 ±5.3) and ATP levels (10.6 ±1.1 and 15.5 ±1.14) compared to MPTP (60.1 ±3.9 pmol/g and 3.6 ±0.09 mmol/g, respectively) (p < 0.05). Moreover, mitochondrial DNA from mice treated with levodopa or stem cells was in intact form; average concentration was (52.8 ±3.01 and 107.8 ±8.6) and no appreciable fragmentation of nuclear DNA was found compared to MPTP group. Regarding tyrosine hydroxylase (TH) immunostaining, stem cell group showed a marked increase of percentage of TH-immunopositive neurons (63.55 ±5.2) compared to both MPTP (37.6 ±3.1) and levodopa groups (41.6 ±3.5). CONCLUSIONS CD34+ cells ameliorated motor, biochemical and histological deficits in MPTP-parkinsonian mice, these effects were superior to those produced by levodopa that would be promising for the treatment of PD.
Collapse
Affiliation(s)
- Noha Abo-Grisha
- Department of Physiology, Faculty of Medicine, Suez Canal University, Suez, Egypt
| | - Soha Essawy
- Pharmacology Department, Faculty of Medicine, Suez Canal University, Suez, Egypt
| | - Dina M. Abo-Elmatty
- Department of Biochemistry, Faculty of Pharmacy, Suez Canal University, Suez, Egypt
| | - Zenab Abdel-Hady
- Department of Histology, Faculty of Medicine, Suez Canal University, Suez, Egypt
| |
Collapse
|
18
|
Liu X, Ye R, Yan T, Yu SP, Wei L, Xu G, Fan X, Jiang Y, Stetler RA, Liu G, Chen J. Cell based therapies for ischemic stroke: from basic science to bedside. Prog Neurobiol 2013; 115:92-115. [PMID: 24333397 DOI: 10.1016/j.pneurobio.2013.11.007] [Citation(s) in RCA: 143] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2013] [Revised: 11/11/2013] [Accepted: 11/26/2013] [Indexed: 12/20/2022]
Abstract
Cell therapy is emerging as a viable therapy to restore neurological function after stroke. Many types of stem/progenitor cells from different sources have been explored for their feasibility and efficacy for the treatment of stroke. Transplanted cells not only have the potential to replace the lost circuitry, but also produce growth and trophic factors, or stimulate the release of such factors from host brain cells, thereby enhancing endogenous brain repair processes. Although stem/progenitor cells have shown a promising role in ischemic stroke in experimental studies as well as initial clinical pilot studies, cellular therapy is still at an early stage in humans. Many critical issues need to be addressed including the therapeutic time window, cell type selection, delivery route, and in vivo monitoring of their migration pattern. This review attempts to provide a comprehensive synopsis of preclinical evidence and clinical experience of various donor cell types, their restorative mechanisms, delivery routes, imaging strategies, future prospects and challenges for translating cell therapies as a neurorestorative regimen in clinical applications.
Collapse
Affiliation(s)
- Xinfeng Liu
- Department of Neurology, Jinling Hospital, Nanjing University School of Medicine, Nanjing, China.
| | - Ruidong Ye
- Department of Neurology, Jinling Hospital, Nanjing University School of Medicine, Nanjing, China
| | - Tao Yan
- Department of Neurology, Henry Ford Hospital, Detroit, MI, USA; Department of Neurology, Tianjin General Hospital, Tianjin University School of Medicine, Tianjin, China
| | - Shan Ping Yu
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA, USA
| | - Ling Wei
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA, USA
| | - Gelin Xu
- Department of Neurology, Jinling Hospital, Nanjing University School of Medicine, Nanjing, China
| | - Xinying Fan
- Department of Neurology, Jinling Hospital, Nanjing University School of Medicine, Nanjing, China
| | - Yongjun Jiang
- Department of Neurology, Jinling Hospital, Nanjing University School of Medicine, Nanjing, China
| | - R Anne Stetler
- Center of Cerebrovascular Disease Research, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - George Liu
- Institute of Cardiovascular Sciences, Peking University Health Science Center, Beijing, China
| | - Jieli Chen
- Department of Neurology, Henry Ford Hospital, Detroit, MI, USA.
| |
Collapse
|
19
|
Ström JO, Ingberg E, Theodorsson A, Theodorsson E. Method parameters' impact on mortality and variability in rat stroke experiments: a meta-analysis. BMC Neurosci 2013; 14:41. [PMID: 23548160 PMCID: PMC3637133 DOI: 10.1186/1471-2202-14-41] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2012] [Accepted: 03/22/2013] [Indexed: 12/14/2022] Open
Abstract
Background Even though more than 600 stroke treatments have been shown effective in preclinical studies, clinically proven treatment alternatives for cerebral infarction remain scarce. Amongst the reasons for the discrepancy may be methodological shortcomings, such as high mortality and outcome variability, in the preclinical studies. A common approach in animal stroke experiments is that A) focal cerebral ischemia is inflicted, B) some type of treatment is administered and C) the infarct sizes are assessed. However, within this paradigm, the researcher has to make numerous methodological decisions, including choosing rat strain and type of surgical procedure. Even though a few studies have attempted to address the questions experimentally, a lack of consensus regarding the optimal methodology remains. Methods We therefore meta-analyzed data from 502 control groups described in 346 articles to find out how rat strain, procedure for causing focal cerebral ischemia and the type of filament coating affected mortality and infarct size variability. Results The Wistar strain and intraluminal filament procedure using a silicone coated filament was found optimal in lowering infarct size variability. The direct and endothelin methods rendered lower mortality rate, whereas the embolus method increased it compared to the filament method. Conclusions The current article provides means for researchers to adjust their middle cerebral artery occlusion (MCAo) protocols to minimize infarct size variability and mortality.
Collapse
Affiliation(s)
- Jakob O Ström
- Department of Clinical and Experimental Medicine, Clinical Chemistry, Faculty of Health Sciences, Linköping University, County Council of Östergötland, Linköping, Sweden.
| | | | | | | |
Collapse
|
20
|
Immune effects of optimized DNA vaccine and protective effects in a MPTP model of Parkinson’s disease. Neurol Sci 2013; 34:1559-70. [DOI: 10.1007/s10072-012-1284-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2012] [Accepted: 12/17/2012] [Indexed: 11/25/2022]
|
21
|
Tajiri N, Acosta S, Glover LE, Bickford PC, Jacotte Simancas A, Yasuhara T, Date I, Solomita MA, Antonucci I, Stuppia L, Kaneko Y, Borlongan CV. Intravenous grafts of amniotic fluid-derived stem cells induce endogenous cell proliferation and attenuate behavioral deficits in ischemic stroke rats. PLoS One 2012; 7:e43779. [PMID: 22912905 PMCID: PMC3422299 DOI: 10.1371/journal.pone.0043779] [Citation(s) in RCA: 73] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2012] [Accepted: 07/25/2012] [Indexed: 01/11/2023] Open
Abstract
We recently reported isolation of viable rat amniotic fluid-derived stem (AFS) cells [1]. Here, we tested the therapeutic benefits of AFS cells in a rodent model of ischemic stroke. Adult male Sprague-Dawley rats received a 60-minute middle cerebral artery occlusion (MCAo). Thirty-five days later, animals exhibiting significant motor deficits received intravenous transplants of rat AFS cells or vehicle. At days 60–63 post-MCAo, significant recovery of motor and cognitive function was seen in stroke animals transplanted with AFS cells compared to vehicle-infused stroke animals. Infarct volume, as revealed by hematoxylin and eosin (H&E) staining, was significantly reduced, coupled with significant increments in the cell proliferation marker, Ki67, and the neuronal marker, MAP2, in the dentate gyrus (DG) [2] and the subventricular zone (SVZ) of AFS cell-transplanted stroke animals compared to vehicle-infused stroke animals. A significantly higher number of double-labeled Ki67/MAP2-positive cells and a similar trend towards increased Ki67/MAP2 double-labeling were observed in the DG and SVZ of AFS cell-transplanted stroke animals, respectively, compared to vehicle-infused stroke animals. This study reports the therapeutic potential of AFS cell transplantation in stroke animals, possibly via enhancement of endogenous repair mechanisms.
Collapse
Affiliation(s)
- Naoki Tajiri
- Department of Neurosurgery and Brain Repair, Center of Excellence for Aging and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, Florida, United States of America
| | - Sandra Acosta
- Department of Neurosurgery and Brain Repair, Center of Excellence for Aging and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, Florida, United States of America
| | - Loren E. Glover
- Department of Neurosurgery and Brain Repair, Center of Excellence for Aging and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, Florida, United States of America
| | - Paula C. Bickford
- Department of Neurosurgery and Brain Repair, Center of Excellence for Aging and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, Florida, United States of America
| | - Alejandra Jacotte Simancas
- Departamento de Psicobiologia y Metodologia de las Cièncias de la Salud, Universidad Autónoma de Barcelona, Barcelona, Spain
| | - Takao Yasuhara
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Isao Date
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Marianna A. Solomita
- Department of Neurosurgery and Brain Repair, Center of Excellence for Aging and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, Florida, United States of America
- Department of Biomedical Sciences, G. d'Annunzio University, Chieti-Pescara, Italy
- Department of Neuroscience and Imaging, School of Advanced Studies G.d'Annunzio, Chieti University and Stem TeCh Group, Aging Research Center, Chieti- Pescara, Italy
| | - Ivana Antonucci
- Department of Biomedical Sciences, G. d'Annunzio University, Chieti-Pescara, Italy
- Department of Neuroscience and Imaging, School of Advanced Studies G.d'Annunzio, Chieti University and Stem TeCh Group, Aging Research Center, Chieti- Pescara, Italy
| | - Liborio Stuppia
- Department of Biomedical Sciences, G. d'Annunzio University, Chieti-Pescara, Italy
- Department of Neuroscience and Imaging, School of Advanced Studies G.d'Annunzio, Chieti University and Stem TeCh Group, Aging Research Center, Chieti- Pescara, Italy
| | - Yuji Kaneko
- Department of Neurosurgery and Brain Repair, Center of Excellence for Aging and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, Florida, United States of America
| | - Cesar V. Borlongan
- Department of Neurosurgery and Brain Repair, Center of Excellence for Aging and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, Florida, United States of America
- * E-mail:
| |
Collapse
|
22
|
Abstract
The glial cell line-derived neurotrophic factor (GDNF) was first identified as a survival factor for midbrain dopaminergic neurons, but additional studies provided evidences for a role as a trophic factor for other neurons of the central and peripheral nervous systems. GDNF regulates cellular activity through interaction with glycosyl-phosphatidylinositol-anchored cell surface receptors, GDNF family receptor-α1, which might signal through the transmembrane Ret tyrosine receptors or the neural cell adhesion molecule, to promote cell survival, neurite outgrowth, and synaptogenesis. The neuroprotective effect of exogenous GDNF has been shown in different experimental models of focal and global brain ischemia, by local administration of the trophic factor, using viral vectors carrying the GDNF gene and by transplantation of GDNF-expressing cells. These different strategies and the mechanisms contributing to neuroprotection by GDNF are discussed in this review. Importantly, neuroprotection by GDNF was observed even when administered after the ischemic injury.
Collapse
Affiliation(s)
- Emília P Duarte
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, Largo Marquês de Pombal, Coimbra, Portugal
| | | | | | | |
Collapse
|
23
|
Tao J, Ji F, Liu B, Wang F, Dong F, Zhu Y. Improvement of deficits by transplantation of lentiviral vector-modified human amniotic mesenchymal cells after cerebral ischemia in rats. Brain Res 2012; 1448:1-10. [PMID: 22386515 DOI: 10.1016/j.brainres.2012.01.069] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2011] [Revised: 01/16/2012] [Accepted: 01/28/2012] [Indexed: 10/14/2022]
Abstract
Amniotic membrane is known to have the ability to transdifferentiate into multiple organs and is expected to stimulate a reduced immunologic reaction. Human amniotic membrane-derived mesenchymal stem cells (hAMCs) do not express the major histocompatibility complex (MHC) class I molecule and may be expected to show immunologic tolerance. A good deal of research has explored the clinical therapeutic potential of hAMCs. In the present study, we isolated hAMCs and transfected them with the brain derived neurotrophic factor (BDNF) gene using lentiviral vectors. These cells were then transplanted into the brains of rats subjected to a transient middle cerebral artery occlusion (MCAO). The hAMCs survived for three weeks in the brains of the ischemic rats, and some of the transplanted hAMCs expressed the neuronal marker MAP2 and the neuronal progenitor marker Nestin. Furthermore, caspase-3 activity and iNOS expression were decreased in the vicinity of the graft and injection site. Importantly, intracerebral grafting of EGFP-modified hAMCs and BDNF-transduced hAMCs significantly ameliorated behavioral dysfunction in ischemic rats. BDNF-hAMCs ameliorated the behavioral dysfunction of rats more rapidly and effectively relative to EGFP-hAMC-treated rats. Finally, the grafts also reduced the infarct volume. hAMCs survived in the brain tissue and improved functional recovery. Because of the lack of ethical concerns and the high supply of these cells, hAMCs represent a promising clinical treatment for gene delivery similar to stem cell strategies.
Collapse
Affiliation(s)
- Jiang Tao
- Department of General Dentistry, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Stomatology, Shanghai, China
| | | | | | | | | | | |
Collapse
|
24
|
Yamagata K. Pathological alterations of astrocytes in stroke-prone spontaneously hypertensive rats under ischemic conditions. Neurochem Int 2011; 60:91-8. [PMID: 22100568 DOI: 10.1016/j.neuint.2011.11.002] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2011] [Revised: 10/27/2011] [Accepted: 11/04/2011] [Indexed: 11/18/2022]
Abstract
Stroke-prone spontaneously hypertensive rats (SHRSP/Izm) develop severe hypertension, and more than 95% of them die of cerebral stroke. We showed the vulnerability of neuronal cells of SHRSP/Izm rats. Furthermore, we analyzed the characteristics of SHRSP/Izm astrocytes during a stroke. It is known that the proliferating ability of SHRSP/Izm astrocytes is significantly enhanced compared with those in the normotensive Wistar Kyoto rats (WKY/Izm) strain. Conversely, the ability of SHRSP/Izm astrocytes to form tight junctions (TJ) was attenuated compared with astrocytes from WKY/Izm rats. During the stress of hypoxia and reoxygenation (H/R), lactate production, an energy source for neuronal cells, decreased in SHRSP/Izm astrocytes in comparison with the WKY/Izm strain. Moreover, during H/R, SHRSP/Izm astrocytes decreased their production of glial cell line-derived neurotrophic factor (GDNF) in comparison with WKY/Izm astrocytes. Furthermore, SHRSP/Izm rats decreased production of l-serine, compared with WKY/Izm rats following nitric oxide (NO) stimulation. Additionally, in H/R, astrocytes of SHRSP/Izm rats expressed adhesion molecules such as VCAM-1 at higher levels. It is possible that all of these differences between SHRSP/Izm and WKY/Izm astrocytes are not associated with the neurological disorders in SHRSP/Izm. However, attenuated production of lactate and reduced GDNF production in astrocytes may reduce required energy levels and weaken the nutritional status of SHRSP/Ism neuronal cells. We suggest that the attenuation of astrocytes' functions accelerates neuronal cell death during stroke, and may contribute to the development of strokes in SHRSP/Izm. In this review, we summarize the altered properties of SHRSP/Izm astrocytes during a stroke.
Collapse
Affiliation(s)
- Kazuo Yamagata
- Laboratory of Molecular Health Science of Food, Department of Food Bioscience and Biotechnology, College of Bioresource Sciences, Nihon University (NUBS), 1866 Kameino, Fujisawa-shi, Kanagawa 252-8510, Japan.
| |
Collapse
|
25
|
Boltze J, Reich DM, Hau S, Reymann KG, Strassburger M, Lobsien D, Wagner DC, Kamprad M, Stahl T. Assessment of neuroprotective effects of human umbilical cord blood mononuclear cell subpopulations in vitro and in vivo. Cell Transplant 2011; 21:723-37. [PMID: 21929866 DOI: 10.3727/096368911x586783] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Experimental transplantation of human umbilical cord blood (hUCB) mononuclear cells (MNCs) in rodent stroke models revealed the therapeutic potential of these cells. However, effective cells within the heterogeneous MNC population and their modes of action are still under discussion. MNCs and MNC fractions enriched (CD34(+)) or depleted (CD34(-)) for CD34-expressing stem/progenitor cells were isolated from hUCB. Cells were transplanted intravenously following middle cerebral artery occlusion in spontaneously hypertensive rats and directly or indirectly cocultivated with hippocampal slices previously subjected to oxygen and glucose deprivation. Application of saline solution or a human T-cell line served as controls. In vivo, MNCs, CD34(+) and CD34(-) cells reduced neurofunctional deficits and diminished lesion volume as determined by magnetic resonance imaging. MNCs were superior to other fractions. However, human cells could not be identified in brain tissue 29 days after stroke induction. Following direct application on postischemic hippocampal slices, MNCs reduced neural damage throughout a 3-day observation period. CD34(+) cells provided transient protection for 2 days. The CD34(-) fraction, in contrast to in vivo results, failed to reduce neural damage. Direct cocultivation of MNCs was superior to indirect cocultivation of equal cell numbers. Indirect application of up to 10-fold MNC concentrations enhanced neuroprotection to a level comparable to direct cocultivation. After direct application, MNCs migrated into the slices. Flow cytometric analysis of migrated cells revealed that the CD34(+) cells within MNCs were preferably attracted by damaged hippocampal tissue. Our study suggests that MNCs provide the most prominent neuroprotective effect, with CD34(+) cells seeming to be particularly involved in the protective action of MNCs. CD34(+) cells preferentially home to neural tissue in vitro, but are not superior concerning the overall effect, implying that there is another, still undiscovered, protective cell population. Furthermore, MNCs did not survive in the ischemic brain for longer periods without immunosuppression.
Collapse
Affiliation(s)
- Johannes Boltze
- Fraunhofer Institute for Cell Therapy and Immunology, Leipzig, Germany.
| | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Intravascular Stem Cell Transplantation for Stroke. Transl Stroke Res 2011; 2:250-65. [DOI: 10.1007/s12975-011-0093-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2011] [Accepted: 07/13/2011] [Indexed: 01/01/2023]
|
27
|
Abstract
AbstractThe young human brain is highly plastic and thus early brain lesions can lead to aberrant development of connectivity and mapping of functions. This is why initially in cerebral palsy only subtle changes in spontaneous movements are seen after the time of lesion, followed by a progressive evolution of a movement disorder over many months and years. Thus we propose that interventions to treat cerebral palsy should be initiated as soon as possible in order to restore the nervous system to the correct developmental trajectory. One such treatment might be autologous stem cell transplantation either intracerebrally or intravenously. All babies come with an accessible supply of stem cells, the umbilical cord, which can supply cells that could theoretically replace missing neural cell types, or act indirectly by supplying trophic support or modulating inflammatory responses to hypoxia/ischaemia. However, for such radical treatment to be proposed, it is necessary to be able to detect and accurately predict the outcomes of brain injury from a very early age. This article reviews our current understanding of perinatal injuries that lead to cerebral palsy, how well modern imaging might predict outcomes, what stem cells are yielded from umbilical cord blood and experimental models of brain repair using stem cells.
Collapse
|