1
|
Sharma H, Reeta KH, Sharma U, Suri V. Decanoic acid mitigates ischemia reperfusion injury by modulating neuroprotective, inflammatory and oxidative pathways in middle cerebral artery occlusion model of stroke in rats. J Stroke Cerebrovasc Dis 2023; 32:107184. [PMID: 37276786 DOI: 10.1016/j.jstrokecerebrovasdis.2023.107184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 05/06/2023] [Accepted: 05/09/2023] [Indexed: 06/07/2023] Open
Abstract
OBJECTIVE Amino-3-hydroxy-5-methyl-4-isoxazole propionate (AMPA) is an ionotropic transmembrane receptor for glutamate. AMPA receptor blockers have been reported to prevent neurological damage and enhance the post stroke recovery in rats. Decanoic acid, a medium-chain fatty acid, has been reported to exhibit non-competitive AMPA receptor antagonism. This study evaluated the effect of decanoic acid administered before and after ischemia reperfusion injury on neurological damage and post stroke recovery in rats. METHODS Middle cerebral artery occlusion (MCAo) was performed by using the intraluminal method to induce focal cerebral ischemia. Decanoic acid (120 mg/kg) was administered orally for 1 day (5-10 min post reperfusion) in one group and for 2 days (24 h pre and 5-10 min post reperfusion) in the other group. Effect on neurological damage and post stroke recovery was assessed by neurobehavioral parameters, MRI and TTC staining along with inflammatory, oxidative, apoptotic, and neuroprotective biomarkers. RESULTS Decanoic acid significantly reduced the MCAo induced neurological damage and infarct size. Decanoic acid treatment increased the motor coordination and grip strength. Furthermore, levels of inflammatory (TNFα, IL-1β and IL-6), oxidative stress (MDA), apoptotic (TUNEL positive cells) and neurological injury (GFAP) biomarkers were reduced after decanoic acid treatment. Anti-inflammatory cytokine (IL-10) and neuroprotective markers (NT-3, BDNF and TrkB) were found to be significantly increased with decanoic acid treatment. CONCLUSION This study showed protective effects of decanoic acid against ischemia reperfusion injury in rats. Anti-inflammatory, antioxidant, neuroprotective, and anti-apoptotic properties may be responsible for the beneficial effects of decanoic acid observed in the study.
Collapse
Affiliation(s)
- Himanshu Sharma
- Department of Pharmacology, All India Institute of Medical Sciences, New Delhi, India
| | - K H Reeta
- Department of Pharmacology, All India Institute of Medical Sciences, New Delhi, India.
| | - Uma Sharma
- Department of NMR, All India Institute of Medical Sciences, New Delhi, India
| | - Vaishali Suri
- Department of Pathology, All India Institute of Medical Sciences, New Delhi, India
| |
Collapse
|
2
|
Niu Y, Du SZ, He R. TNF-α interference ameliorates brain damage in neonatal hypoxic-ischemic encephalopathy rats by regulating the expression of NT-3 and TRKC. IBRAIN 2023; 9:381-389. [PMID: 38680513 PMCID: PMC11045181 DOI: 10.1002/ibra.12089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 01/06/2023] [Accepted: 01/13/2023] [Indexed: 05/01/2024]
Abstract
The aim of this study is to explore the effect of tumor necrosis factor-α (TNF-α) inhibition in rats with neonatal hypoxic-ischemic encephalopathy (HIE) and ascertain the relevant signaling pathways. The Zea-Longa score was used to evaluate the neurological function of the rats. ImageJ was used for quantification of the brain edema volume. Triphenyl tetrazolium chloride (TTC) staining of brain tissue was performed 24 h after hypoxic-ischemic (HI) to detect right brain infarction. The expression of TNF-α was detected by real-time quantitative polymerase chain reaction (RT-qPCR). Immunofluorescence staining was used to identify the localization of TNF-α; Then, the effective shRNA fragment of TNF-α was used to validate the role of TNF-α in HIE rats, and the change of neurotrofin-3 (NT-3) and tyrosine kinase receptor-C (TRKC) was examined after TNF-α-shRNA lentivirus transfection to determine downstream signaling associated with TNF-α. Protein interaction analysis was carried out to predict the links among TNF-α, NT-3, and TRKC. Cerebral edema volume and infarction increased in the right brain after the HI operation. The Zea-Longa score significantly increased within 24 h after the HI operation. The relative expression of TNF-α was upregulated after the HI operation. TNF-α was highly expressed in the right hippocampus post HI through immunofluorescence staining. Bioinformatics analysis found a direct or an indirect link among TNF-α, NT-3, and TRKC. Moreover, the interference of TNF-α increased the expression of NT-3 and TRKC. TNF-α interference might alleviate brain injury in HIE by upregulating NT-3 and TRKC.
Collapse
Affiliation(s)
- Yong‐Min Niu
- Institute of NeuroscienceKunming Medical UniversityKunmingChina
| | - Steven Z. Du
- Department of Integrative BiologyUniversity of Wisconsin‐MadisonMadisonWisconsinUSA
| | - Rong He
- Animal Zoology DepartmentKunming Medical UniversityKunmingChina
| |
Collapse
|
3
|
Neuroprotection of Oral Edaravone on Middle Cerebral Artery Occlusion in Rats. Neurotox Res 2022; 40:995-1006. [DOI: 10.1007/s12640-022-00520-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 05/13/2022] [Accepted: 05/13/2022] [Indexed: 10/18/2022]
|
4
|
Omar NA, Kumar J, Teoh SL. Neurotrophin-3 and neurotrophin-4: The unsung heroes that lies behind the meninges. Neuropeptides 2022; 92:102226. [PMID: 35030377 DOI: 10.1016/j.npep.2022.102226] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 12/06/2021] [Accepted: 01/04/2022] [Indexed: 11/17/2022]
Abstract
Neurotrophin is a growth factor that regulates the development and repair of the nervous system. From all factors, two pioneer groups, the nerve growth factor (NGF) and brain derived neurotrophic factor (BDNF), have been widely explored for their role in disease pathogenesis and potential use as therapeutic agents. Nonetheless, neurotrophin-3 (NT3) and neurotrophin-4 (NT4) also have promising potential, albeit less popular than their counterparts. This review focuses on the latter two factors and their roles in the pathogenesis of brain disorders and potential therapies. An extensive literature search of NT3 and NT4 with their receptors, the TrkB and TrkC on the nervous system were extracted and analyzed. We found that NT3 and NT4 are not only involved in the pathogenesis of some neurodegenerative diseases, but also have promising therapeutic potential on injury- and vascular-related nervous system disease, neuropsychiatry, neurodegeneration and peripheral nerve diseases. In conclusion, the role of NT3 and NT4 should be further emphasized, and more studies could be explored on the potential use of these neurotrophins in the human study.
Collapse
Affiliation(s)
- Noor Azzizah Omar
- Department of Anatomy, Universiti Kebangsaan Malaysia Medical Centre, Bandar Tun Razak, 56000 Kuala Lumpur, Malaysia; Department of Medical Sciences, Faculty of Medicine and Health Sciences, Universiti Sains Islam Malaysia, 71800 Nilai, Negeri Sembilan, Malaysia.
| | - Jaya Kumar
- Department of Physiology, Universiti Kebangsaan Malaysia Medical Centre, Bandar Tun Razak, 56000 Kuala Lumpur, Malaysia.
| | - Seong Lin Teoh
- Department of Anatomy, Universiti Kebangsaan Malaysia Medical Centre, Bandar Tun Razak, 56000 Kuala Lumpur, Malaysia.
| |
Collapse
|
5
|
Results from the Population-Based Gutenberg Health Study Revealing Four Altered Autoantibodies in Retinal Vein Occlusion Patients. J Ophthalmol 2020; 2020:8386160. [PMID: 32802490 PMCID: PMC7411451 DOI: 10.1155/2020/8386160] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2020] [Accepted: 06/30/2020] [Indexed: 12/22/2022] Open
Abstract
Purpose Retinal vein occlusion (RVO) is the second most common retinal vascular disease and a major cause of visual impairment. In this study, we aimed to observe whether RVO cases have different antibody profiles as a new potential risk factor and whether a conversion of retinal vein occlusion (RVO) to neovascular glaucoma (NVG), one of the major complications, is occurring within a 5-year timeframe. Methods We performed a nested case-control study (1 : 4) within the Gutenberg Health Study (GHS), a population-based, prospective cohort study in the Rhine-Main Region of Germany including 15,010 participants. RVO subjects (n = 59) were identified by grading of fundus photographs. Optic nerves of RVO subjects and age- and sex-matched controls (n = 229) at baseline and their follow-up examination after 5 years were analyzed for glaucomatous alterations. Of all RVO subjects and controls, serum autoantibody profiles were measured using in-house manufactured antigen-antibody microarrays. Results Of the 59 RVO patients, 3 patients (5%) showed glaucomatous optic disc alterations at baseline, whereas no new glaucoma case was detected at 5-year follow-up. Four of the autoantibodies measured (against dermcidin, neurotrophin-3, superoxide dismutase 1, and signal recognition particle 14 kDa protein) were significantly increased in the serum of RVO patients (p < 0.001). Multivariable conditional logistic regression analysis showed that 3 of these 4 antibodies were independent of cardiovascular risk factors. Conclusions We found several autoantibodies associated with RVO, targeting proteins and structures possibly involved in RVO pathogenesis.
Collapse
|
6
|
TNF-α Pretreatment Improves the Survival and Function of Transplanted Human Neural Progenitor Cells Following Hypoxic-Ischemic Brain Injury. Cells 2020; 9:cells9051195. [PMID: 32403417 PMCID: PMC7291333 DOI: 10.3390/cells9051195] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Revised: 04/28/2020] [Accepted: 05/07/2020] [Indexed: 02/07/2023] Open
Abstract
Neural progenitor cells (NPCs) therapy offers great promise in hypoxic-ischemic (HI) brain injury. However, the poor survival of implanted NPCs in the HI host environment limits their therapeutic effects. Tumor necrosis factor-alpha (TNF-α) is a pleiotropic cytokine that is induced in response to a variety of pathological processes including inflammation and immunity. On the other hand, TNF-α has protective effects on cell apoptosis and death and affects the differentiation, proliferation, and survival of neural stem/progenitor cells in the brain. The present study investigated whether TNF-α pretreatment on human NPCs (hNPCs) enhances the effectiveness of cell transplantation therapy under ischemic brain. Fetal brain tissue-derived hNPCs were pretreated with TNF-α before being used in vitro experiments or transplantation. TNF-α significantly increased expression of cIAP2, and the use of short hairpin RNA-mediated knockdown of cIAP2 demonstrated that cIAP2 protected hNPCs against HI-induced cytotoxicity. In addition, pretreatment of hNPCs with TNF-α mediated neuroprotection by altering microglia polarization via increased expression of CX3CL1 and by enhancing expression of neurotrophic factors. Furthermore, transplantation of TNF-α-treated hNPCs reduced infarct volume and improved neurological functions in comparison with non-pretreated hNPCs or vehicle. These findings show that TNF-α pretreatment, which protects hNPCs from HI-injured brain-induced apoptosis and increases neuroprotection, is a simple and safe approach to improve the survival of transplanted hNPCs and the therapeutic efficacy of hNPCs in HI brain injury.
Collapse
|
7
|
Barui AK, Jhelum P, Nethi SK, Das T, Bhattacharya D, B V, Karri S, Chakravarty S, Patra CR. Potential Therapeutic Application of Zinc Oxide Nanoflowers in the Cerebral Ischemia Rat Model through Neuritogenic and Neuroprotective Properties. Bioconjug Chem 2020; 31:895-906. [PMID: 32050064 DOI: 10.1021/acs.bioconjchem.0c00030] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Neuritogenesis, a complex process of the sprouting of neurites, plays a vital role in the structural and functional restoration of cerebral ischemia-injured neuronal tissue. Practically, there is no effective long-term treatment strategy for cerebral ischemia in clinical practice to date due to several limitations of conventional therapies, facilitating the urgency to develop new alternative therapeutic approaches. Herein, for the first time we report that pro-angiogenic nanomaterials, zinc oxide nanoflowers (ZONF), exhibit neuritogenic activity by elevating mRNA expression of different neurotrophins, following PI3K/Akt-MAPK/ERK signaling pathways. Further, ZONF administration to global cerebral ischemia-induced Fischer rats shows improved neurobehavior and enhanced synaptic plasticity of neurons via upregulation of Neurabin-2 and NT-3, revealing their neuroprotective activity. Altogether, this study offers the basis for exploitation of angio-neural cross talk of other pro-angiogenic nano/biomaterials for future advancement of alternative treatment strategies for cerebral ischemia, where neuritogenesis and neural repair are highly critical.
Collapse
Affiliation(s)
- Ayan Kumar Barui
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology, Uppal Road, Tarnaka, Hyderabad 500007, India.,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh 201002, India
| | - Priya Jhelum
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology, Uppal Road, Tarnaka, Hyderabad 500007, India
| | - Susheel Kumar Nethi
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology, Uppal Road, Tarnaka, Hyderabad 500007, India.,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh 201002, India
| | - Tapatee Das
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology, Uppal Road, Tarnaka, Hyderabad 500007, India.,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh 201002, India
| | - Dwaipayan Bhattacharya
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology, Uppal Road, Tarnaka, Hyderabad 500007, India
| | - Vinothkumar B
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology, Uppal Road, Tarnaka, Hyderabad 500007, India
| | - Shailaja Karri
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology, Uppal Road, Tarnaka, Hyderabad 500007, India
| | - Sumana Chakravarty
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology, Uppal Road, Tarnaka, Hyderabad 500007, India.,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh 201002, India
| | - Chitta Ranjan Patra
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology, Uppal Road, Tarnaka, Hyderabad 500007, India.,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh 201002, India
| |
Collapse
|
8
|
Ghali GZ, Ghali MGZ. Nafamostat mesylate attenuates the pathophysiologic sequelae of neurovascular ischemia. Neural Regen Res 2020; 15:2217-2234. [PMID: 32594033 PMCID: PMC7749469 DOI: 10.4103/1673-5374.284981] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Nafamostat mesylate, an apparent soi-disant panacea of sorts, is widely used to anticoagulate patients undergoing hemodialysis or cardiopulmonary bypass, mitigate the inflammatory response in patients diagnosed with acute pancreatitis, and reverse the coagulopathy of patients experiencing the commonly preterminal disseminated intravascular coagulation in the Far East. The serine protease inhibitor nafamostat mesylate exhibits significant neuroprotective effects in the setting of neurovascular ischemia. Nafamostat mesylate generates neuroprotective effects by attenuating the enzymatic activity of serine proteases, neuroinflammatory signaling cascades, and the endoplasmic reticulum stress responses, downregulating excitotoxic transient receptor membrane channel subfamily 7 cationic currents, modulating the activity of intracellular signal transduction pathways, and supporting neuronal survival (brain-derived neurotrophic factor/TrkB/ERK1/2/CREB, nuclear factor kappa B. The effects collectively reduce neuronal necrosis and apoptosis and prevent ischemia mediated disruption of blood-brain barrier microarchitecture. Investigational clinical applications of these compounds may mitigate ischemic reperfusion injury in patients undergoing cardiac, hepatic, renal, or intestinal transplant, preventing allograft rejection, and treating solid organ malignancies. Neuroprotective effects mediated by nafamostat mesylate support the wise conduct of randomized prospective controlled trials in Western countries to evaluate the clinical utility of this compound.
Collapse
Affiliation(s)
- George Zaki Ghali
- United States Environmental Protection Agency, Arlington, VA; Department of Toxicology, Purdue University, West Lafayette, IN, USA
| | - Michael George Zaki Ghali
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA; Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA, USA
| |
Collapse
|
9
|
Wang Q, Wei J, Shi Y. Platelet microvesicles promote the recovery of neurological function in mouse model of cerebral infarction by inducing angiogenesis. Biochem Biophys Res Commun 2019; 513:997-1004. [PMID: 31005253 DOI: 10.1016/j.bbrc.2019.04.083] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Accepted: 04/11/2019] [Indexed: 12/17/2022]
Abstract
The aim of this study is to investigate the effect of PMVs on mice with ischemic cerebral infarction and its mechanism. Male C57BL/6 mice were selected, and the right focal cortical infarction model was established via cauterization under a microscope and randomly divided into sham operation (Sham) group, normal saline control (Saline) group and platelet microvesicles intervention (PMVs) group. At 1 h after modeling, 5 μL of PMVs (50 μg/mL) or normal saline was injected into the lateral ventricle. The neurological function of mice in each group was evaluated at 1, 3, 7, 14 and 28 d after modeling. After 28 d, the cerebral infarction area was detected via 2,3,5-triphenyltetrazolium chloride (TTC) staining. At 7 and 28 d after modeling, the blood vessel density, proliferation rate of new vessels and encapsulation rate of pericytes were detected via immunofluorescence staining. Moreover, the changes in cerebral cortical blood flow at the infarction side were detected before modeling and at 7 and 28 d after modeling, respectively. Finally, the expressions of proangiogenic factors vascular endothelial growth factor (VEGF), angiopoietin-1 (Ang-1) and N-Cadherin were detected via Western blotting at 3, 7 and 28 d after modeling. PMVs could promote the improvement of neurological function and significantly reduce the cerebral infarction volume in mice with cerebral infarction. PMVs promoted proliferation of new vessels and increased blood vessel density at the infarction edge in mice with cerebral infarction. PMVs could increase the encapsulation rate of pericytes at the infarction edge and improve the permeability of blood-brain barrier in mice with cerebral infarction. PMVs could increase the cerebral cortical blood flow perfusion in mice with cerebral infarction. PMVs could increase proangiogenic factors in brain tissues in mice with cerebral infarction. PMVs could significantly improve the recovery of neurological function in mice with cerebral infarction, which is closely related to the ability of PMVs to promote angiogenesis at the infarction edge. The possible mechanism is that PMVs facilitate angiogenesis after cerebral infarction through promoting the expressions of VEGF, Ang-1 and N-Cadherin. More importantly, the new vessels promoted by PMVs have complete structure and perfect function, and can improve the cerebral blood flow perfusion at the infarction side.
Collapse
Affiliation(s)
- Qing Wang
- Department of Neurology, The First Hospital of Xi'an, Xi'an, China
| | - Jiachen Wei
- Department of Endocrinology, The First Hospital of Xi'an, Xi'an, China
| | - Yaling Shi
- Department of Neurology, The First Hospital of Xi'an, Xi'an, China.
| |
Collapse
|
10
|
Sun C, Fu J, Qu Z, Li D, Si P, Qiao Q, Zhang W, Xue Y, Zhen J, Wang W. Chronic mild hypoxia promotes hippocampal neurogenesis involving Notch1 signaling in epileptic rats. Brain Res 2019; 1714:88-98. [PMID: 30768929 DOI: 10.1016/j.brainres.2019.02.011] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Revised: 02/06/2019] [Accepted: 02/11/2019] [Indexed: 01/08/2023]
Abstract
Cognitive impairment is one of the most common and disabling co-morbidities of epilepsy. It is therefore imperative to find novel treatment approaches to rescue cognitive function among epilepsy patients. Adult neurogenesis is strongly implicated in cognitive function, and mild hypoxia is known to promote the proliferation and differentiation of both embryonic and adult neural stem cells (NSCs). In the present study, we investigated the effect of mild hypoxia on cognitive function and hippocampal neurogenesis of rats with pilocarpine-induced chronic epilepsy. Chronic epilepsy induced marked spatial learning and memory deficits in the Morris water maze that were rescued by consecutively 28 days mild hypoxia exposure (6 h/d at 3000 m altitude equivalent) during the chronic phase. Moreover, mild hypoxia reversed the suppression of hippocampal neurogenesis and the downregulation of NT-3 and BDNF expression in hippocampus and cortex of epileptic rats. Mild hypoxia in vitro also promoted hippocampus-derived NSC proliferation and neuronal differentiation. In addition, mild hypoxia enhanced Notch1 and Hes1 expression, suggesting that Notch1 signaling may be involved in neuroprotection of hypoxia. Our data may help to pave the way for identifying new therapeutic targets for rescuing cognition conflicts in epileptic patients by using hypoxia to promote hippocampus neurogenesis.
Collapse
Affiliation(s)
- Can Sun
- Department of Neurology, Key Laboratory of Neurology of Hebei Province, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, People's Republic of China
| | - Jian Fu
- Department of Emergency, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, People's Republic of China
| | - Zhenzhen Qu
- Department of Neurology, Key Laboratory of Neurology of Hebei Province, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, People's Republic of China
| | - Dongxiao Li
- Department of Neurology, Key Laboratory of Neurology of Hebei Province, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, People's Republic of China
| | - Peipei Si
- Department of Neurology, Key Laboratory of Neurology of Hebei Province, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, People's Republic of China
| | - Qi Qiao
- Department of Neurology, Key Laboratory of Neurology of Hebei Province, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, People's Republic of China
| | - Wenlin Zhang
- Department of Neurology, Key Laboratory of Neurology of Hebei Province, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, People's Republic of China
| | - Yan Xue
- Department of Neurology, Key Laboratory of Neurology of Hebei Province, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, People's Republic of China
| | - Junli Zhen
- Department of Neurology, Key Laboratory of Neurology of Hebei Province, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, People's Republic of China.
| | - Weiping Wang
- Department of Neurology, Key Laboratory of Neurology of Hebei Province, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, People's Republic of China.
| |
Collapse
|
11
|
Neural Stem Cell-Conditioned Medium Ameliorated Cerebral Ischemia-Reperfusion Injury in Rats. Stem Cells Int 2018; 2018:4659159. [PMID: 29765412 PMCID: PMC5903322 DOI: 10.1155/2018/4659159] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2017] [Revised: 09/22/2017] [Accepted: 10/10/2017] [Indexed: 01/08/2023] Open
Abstract
Introduction Our previous study suggested that NSC-CM (neural stem cell-conditioned medium) inhibited cell apoptosis in vitro. In addition, many studies have shown that neurotrophic factors and microparticles secreted into a conditioned medium by NSCs had neuroprotective effects. Thus, we hypothesized that NSC-CM had the capacity of protecting against cerebral I/R injury. Methods Adult male Sprague-Dawley rats receiving middle cerebral artery occlusion surgery as an animal model of cerebral I/R injury were randomly assigned to two groups: the control group and NSC-CM-treated group. 1.5 ml NSC-CM or PBS (phosphate buffer saline) was administrated slowly by tail vein at 3 h, 24 h, and 48 h after ischemia onset. Results NSC-CM significantly ameliorated neurological defects and reduced cerebral infarct volume, accompanied by preserved mitochondrial ultrastructure. In addition, we also found that NSC-CM significantly inhibited cell apoptosis in the ischemic hemisphere via improving the expression of Bcl-2 (B-cell lymphoma-2). Conclusion NSC-CM might be an alternative and effective therapeutic intervention for ischemic stroke.
Collapse
|
12
|
Expression of Neurotrophin-3 and trkC following Focal Cerebral Ischemia in Adult Rat Brain with Treadmill Exercise. BIOMED RESEARCH INTERNATIONAL 2017; 2017:9248542. [PMID: 29018822 PMCID: PMC5606098 DOI: 10.1155/2017/9248542] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/23/2016] [Revised: 04/12/2017] [Accepted: 08/01/2017] [Indexed: 01/09/2023]
Abstract
Neurotrophin-3 (NT-3) is a neurotrophic factor that mainly binds to the tyrosine kinase C (trkC) receptor. NT-3 has been shown to have neuroprotective effects in focal cerebral ischemia. Exercise also has ability to induce functional recovery in focal cerebral ischemia. However, the relationship between NT-3, its receptor trkC, and exercise has not been revealed. In this study, we assessed the expressions of NT-3 and trkC in focal cerebral ischemia. We also assessed the expression of NT-3 and trkC with treadmill exercise in focal cerebral ischemia. The results showed that, in a permanent middle cerebral artery occlusion rat model, exercise increased NT-3 and trkC expression. However, the patterns of expression of NT-3 and trkC at different time points varied. These results suggest that exercise-induced functional recovery in focal cerebral ischemia was related to NT-3 and trkC, but the role on times of NT-3 and trkC differed, although trkC is the receptor kinase for NT-3.
Collapse
|
13
|
Dai Y, Li W, Zhong M, Chen J, Cheng Q, Liu Y, Li T. The paracrine effect of cobalt chloride on BMSCs during cognitive function rescue in the HIBD rat. Behav Brain Res 2017; 332:99-109. [PMID: 28576310 DOI: 10.1016/j.bbr.2017.05.055] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2017] [Revised: 05/20/2017] [Accepted: 05/24/2017] [Indexed: 12/22/2022]
Abstract
Hypoxia-ischemia (HI)-induced perinatal encephalopathy frequently causes chronic neurological morbidities and acute mortality. Bone mesenchymal stem cell (BMSC) transplantation could potentially promote functional and anatomical recovery of ischemic tissue. In vitro hypoxic preconditioning is an effective strategy to improve the survival of BMSCs in ischemic tissue. In this study, cobalt chloride (CoCl2) preconditioned medium from BMSC cultures was injected into the left lateral ventricle of HI rats using a micro-osmotic pump at a flow rate 1.0μl/h for 7 days. The protein levels of HIF-1α and its target genes, vascular endothelial growth factor and erythropoietin, markedly increased after CoCl2 preconditioning in BMSCs. In 7-week-old rats that received CoCl2 preconditioned BMSC medium, results of the Morris water maze test indicated ameliorated spatial working memory function following hypoxia-ischemia damage. Neuronal loss, cellular disorganization, and shrinkage in brain tissue were also ameliorated. Extracellular field excitatory postsynaptic potentials (fEPSPs) in the brain slices of 8-week-old rats were recorded; administration of CoCl2 preconditioned BMSC culture medium induced a progressive increment of baseline and amplitude of the fEPSPs. Immunohistochemical quantification showed that GluR2 protein expression increased. In conclusion, CoCl2 activates HIF-1α signals in BMSCs. CoCl2 preconditioned BMSC culture medium likely effects neuroprotection by inducing long-term potentiation (LTP), which could be associated with GluR2 expression. The paracrine effects of hypoxia preconditioning on BMSCs could have applications in novel cell-based therapeutic strategies for hypoxic and ischemic brain injury.
Collapse
Affiliation(s)
- Ying Dai
- Department of Primary Child Health Care, Children's Hospital of Chongqing Medical University, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China
| | - Wendi Li
- Pediatric Research Institute, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing Engineering Research Center of Stem Cell Therapy, China
| | - Min Zhong
- Department of Neurology, Children's Hospital of Chongqing Medical University, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China
| | - Jie Chen
- Pediatric Research Institute, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing Engineering Research Center of Stem Cell Therapy, China
| | - Qian Cheng
- Department of Primary Child Health Care, Children's Hospital of Chongqing Medical University, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China
| | - Youxue Liu
- Pediatric Research Institute, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing Engineering Research Center of Stem Cell Therapy, China.
| | - Tingyu Li
- Department of Primary Child Health Care, Children's Hospital of Chongqing Medical University, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China; Pediatric Research Institute, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing Engineering Research Center of Stem Cell Therapy, China.
| |
Collapse
|
14
|
Darabi S, Tiraihi T, Delshad A, Sadeghizadeh M, Khalil W, Taheri T. In vitro non-viral murine pro-neurotrophin 3 gene transfer into rat bone marrow stromal cells. J Neurol Sci 2017; 375:137-145. [PMID: 28320116 DOI: 10.1016/j.jns.2017.01.058] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2016] [Revised: 01/16/2017] [Accepted: 01/19/2017] [Indexed: 12/12/2022]
Abstract
Neurotrophin 3 (NT-3) is an important factor for promoting prenatal neural development, as well as regeneration, axogenesis and plasticity in postnatal life. Therapy with NT-3 was reported to improve the condition of patients suffering from degenerative diseases and traumatic injuries, however, the disadvantage of NT-3 protein delivery is its short half-life, thus our alternative approach is the use of NT-3 gene therapy. In this study, the bone marrow stromal cells (BMSCs) were isolated from adult rats, cultured for 4 passages and transfected with either pEGFP-N1 or a constructed vector containing murine proNT-3 (pSecTag2/HygroB-murine proNT-3) using Lipofectamine 2000 followed by Hygromycin B (200mg/kg). The transfection efficiency of the transiently transfected BMSCs was evaluated using the green fluorescence protein containing vector (pEGFP-N1). A quantitative evaluation of the NT-3 expression of mRNA using real time qRT-PCR shows that there was double fold increase in NT-3 gene expression compared with non-transfected BMSCs, also, the culture supernatant yielded double fold increase in NT-3 using ELISA technique, the data were supported by immunoblotting technique. This suggests that the use of this transfection technique can be useful for gene therapy in different neurological disorders with neurodegenerative or traumatic origins.
Collapse
Affiliation(s)
- Shahram Darabi
- Cellular and Molecular Research Center, Qazvin University of Medical Science, Qazvin, Iran
| | - Taki Tiraihi
- Department of Anatomical Sciences, School of Medical Sciences, Tarbiat Modares University, Tehran, Iran.
| | | | - Majid Sadeghizadeh
- Department of Genetics, Faculty of Basic Sciences, Tarbiat Modares University, Tehran, Iran
| | - Wisam Khalil
- Department of Anatomical Sciences, School of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Taher Taheri
- Shefa Neurosciences Research Center, Khatam Al-Anbia Hospital, Tehran, Iran
| |
Collapse
|
15
|
Abstract
INTRODUCTION Ischemic stroke is caused by reduced blood supply and leads to loss of brain function. The reduced oxygen and nutrient supply stimulates various physiological responses, including induction of growth factors. Growth factors prevent neuronal cell death, promote neovascularization, and induce cell growth. However, the concentration of growth factors is not sufficient to recover brain function after the ischemic damage, suggesting that delivery of growth factors into the ischemic brain may be a useful treatment for ischemic stroke. AREAS COVERED In this review, various approaches for the delivery of growth factors to ischemic brain tissue are discussed, including local and targeting delivery systems. EXPERT OPINION To develop growth factor therapy for ischemic stroke, important considerations should be taken into account. First, growth factors may have possible side effects. Thus, concentration of growth factors should be restricted to the ischemic tissues by local administration or targeted delivery. Second, the duration of growth factor therapy should be optimized. Growth factor proteins may be degraded too fast to have a high enough therapeutic effect. Therefore, delivery systems for controlled release or gene delivery may be useful. Third, the delivery systems to the brain should be optimized according to the delivery route.
Collapse
Affiliation(s)
- Taiyoun Rhim
- a Department of Bioengineering, College of Engineering , Hanyang University , Seoul , Republic of Korea
| | - Minhyung Lee
- a Department of Bioengineering, College of Engineering , Hanyang University , Seoul , Republic of Korea
| |
Collapse
|
16
|
Intranasal delivery of bFGF with nanoliposomes enhances in vivo neuroprotection and neural injury recovery in a rodent stroke model. J Control Release 2016; 224:165-175. [DOI: 10.1016/j.jconrel.2016.01.017] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2015] [Accepted: 01/11/2016] [Indexed: 01/20/2023]
|
17
|
Stavely R, Robinson AM, Miller S, Boyd R, Sakkal S, Nurgali K. Human adult stem cells derived from adipose tissue and bone marrow attenuate enteric neuropathy in the guinea-pig model of acute colitis. Stem Cell Res Ther 2015; 6:244. [PMID: 26652292 PMCID: PMC4674993 DOI: 10.1186/s13287-015-0231-x] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2015] [Revised: 10/22/2015] [Accepted: 11/09/2015] [Indexed: 02/08/2023] Open
Abstract
Introduction Mesenchymal stem cells (MSCs) have been identified as a viable treatment for inflammatory bowel disease (IBD). MSCs derived from bone marrow (BM-MSCs) have predominated in experimental models whereas the majority of clinical trials have used MSCs derived from adipose tissue (AT-MSCs), thus there is little consensus on the optimal tissue source. The therapeutic efficacies of these MSCs are yet to be compared in context of the underlying dysfunction of the enteric nervous system innervating the gastrointestinal tract concomitant with IBD. This study aims to characterise the in vitro properties of MSCs and compare their in vivo therapeutic potential for the treatment of enteric neuropathy associated with intestinal inflammation. Methods BM-MSCs and AT-MSCs were validated and characterised in vitro. In in vivo experiments, guinea-pigs received either 2,4,6-trinitrobenzene-sulfonate acid (TNBS) for the induction of colitis or sham treatment by enema. MSCs were administered at a dose of 1x106 cells via enema 3 hours after the induction of colitis. Colon tissues were collected 24 and 72 hours after TNBS administration to assess the level of inflammation and damage to the ENS. MSC migration to the myenteric plexus in vivo was elucidated by immunohistochemistry and in vitro using a modified Boyden chamber assay. Results Cells exhibited multipotency and a typical surface immunophenotype for validation as bona fide MSCs. In vitro characterisation revealed distinct differences in growth kinetics, clonogenicity and cell morphology between MSC types. In vivo, BM-MSCs were comparatively more effective than AT-MSCs in attenuating leukocyte infiltration and neuronal loss in the myenteric plexus. MSCs from both sources equally ameliorated body weight loss, gross morphological damage to the colon, changes in the neurochemical coding of neuronal subpopulations and the reduction in density of extrinsic and intrinsic nerve fibres innervating the colon. MSCs from both sources migrated to the myenteric plexus in in vivo colitis and in an in vitro assay. Conclusions These data from in vitro experiments suggest that AT-MSCs are ideal for cellular expansion. However, BM-MSCs were more therapeutic in the treatment of enteric neuropathy and plexitis. These characteristics should be considered when deciding on the MSC tissue source.
Collapse
Affiliation(s)
- Rhian Stavely
- Centre for Chronic Disease, College of Health and Biomedicine, Victoria University, Melbourne, Australia.
| | - Ainsley M Robinson
- Centre for Chronic Disease, College of Health and Biomedicine, Victoria University, Melbourne, Australia.
| | - Sarah Miller
- Centre for Chronic Disease, College of Health and Biomedicine, Victoria University, Melbourne, Australia.
| | - Richard Boyd
- Department of Anatomy and Developmental Biology, Monash University, Melbourne, Australia.
| | - Samy Sakkal
- Centre for Chronic Disease, College of Health and Biomedicine, Victoria University, Melbourne, Australia.
| | - Kulmira Nurgali
- Centre for Chronic Disease, College of Health and Biomedicine, Victoria University, Melbourne, Australia. .,College of Health and Biomedicine, Victoria University, Western Centre for Health Research & Education, 176 Furlong Road, St Albans, 3021, VIC, Australia.
| |
Collapse
|
18
|
Duricki DA, Hutson TH, Kathe C, Soleman S, Gonzalez-Carter D, Petruska JC, Shine HD, Chen Q, Wood TC, Bernanos M, Cash D, Williams SCR, Gage FH, Moon LDF. Delayed intramuscular human neurotrophin-3 improves recovery in adult and elderly rats after stroke. Brain 2015; 139:259-75. [PMID: 26614754 PMCID: PMC4785394 DOI: 10.1093/brain/awv341] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2015] [Accepted: 09/29/2015] [Indexed: 12/11/2022] Open
Abstract
There is an urgent need for a therapy that reverses disability after stroke when initiated in a time frame suitable for the majority of new victims. We show here that intramuscular delivery of neurotrophin-3 (NT3, encoded by NTF3) can induce sensorimotor recovery when treatment is initiated 24 h after stroke. Specifically, in two randomized, blinded preclinical trials, we show improved sensory and locomotor function in adult (6 months) and elderly (18 months) rats treated 24 h following cortical ischaemic stroke with human NT3 delivered using a clinically approved serotype of adeno-associated viral vector (AAV1). Importantly, AAV1-hNT3 was given in a clinically-feasible timeframe using a straightforward, targeted route (injections into disabled forelimb muscles). Magnetic resonance imaging and histology showed that recovery was not due to neuroprotection, as expected given the delayed treatment. Rather, treatment caused corticospinal axons from the less affected hemisphere to sprout in the spinal cord. This treatment is the first gene therapy that reverses disability after stroke when administered intramuscularly in an elderly body. Importantly, phase I and II clinical trials by others show that repeated, peripherally administered high doses of recombinant NT3 are safe and well tolerated in humans with other conditions. This paves the way for NT3 as a therapy for stroke.
Collapse
Affiliation(s)
- Denise A Duricki
- 1 Neurorestoration Group, Wolfson Centre for Age-Related Diseases, King's College London, 16-18 Newcomen Street, London SE1 1UL, UK 2 Centre for Integrative Biology, King's College London, Franklin-Wilkins Building, 150 Stamford Street, London SE1 9NH, UK
| | - Thomas H Hutson
- 1 Neurorestoration Group, Wolfson Centre for Age-Related Diseases, King's College London, 16-18 Newcomen Street, London SE1 1UL, UK 3 Division of Brain Sciences, Department of Medicine, Hammersmith Campus, Imperial College London, London, UK
| | - Claudia Kathe
- 1 Neurorestoration Group, Wolfson Centre for Age-Related Diseases, King's College London, 16-18 Newcomen Street, London SE1 1UL, UK
| | - Sara Soleman
- 1 Neurorestoration Group, Wolfson Centre for Age-Related Diseases, King's College London, 16-18 Newcomen Street, London SE1 1UL, UK 4 John Van Geest Centre for Brain Repair University of Cambridge, The E.D. Adrian Building, Forvie Site, Robinson Way Cambridge, CB2 0PY, UK
| | - Daniel Gonzalez-Carter
- 1 Neurorestoration Group, Wolfson Centre for Age-Related Diseases, King's College London, 16-18 Newcomen Street, London SE1 1UL, UK 3 Division of Brain Sciences, Department of Medicine, Hammersmith Campus, Imperial College London, London, UK
| | - Jeffrey C Petruska
- 5 Department of Anatomical Sciences and Neurobiology, University of Louisville; Kentucky Spinal Cord Injury Research Center, Department of Neurological Surgery, Louisville, Kentucky, USA
| | - H David Shine
- 6 Center for Cell and Gene Therapy, Department of Neuroscience, Alkek Bldg N1130.01, Baylor College of Medicine, One Baylor Plaza, Houston, Texas 77030, USA
| | - Qin Chen
- 6 Center for Cell and Gene Therapy, Department of Neuroscience, Alkek Bldg N1130.01, Baylor College of Medicine, One Baylor Plaza, Houston, Texas 77030, USA
| | - Tobias C Wood
- 7 Neuroimaging Research Group, King's College London, PO42 De Crespigny Park, London, SE5 8AF, UK
| | - Michel Bernanos
- 7 Neuroimaging Research Group, King's College London, PO42 De Crespigny Park, London, SE5 8AF, UK
| | - Diana Cash
- 7 Neuroimaging Research Group, King's College London, PO42 De Crespigny Park, London, SE5 8AF, UK
| | - Steven C R Williams
- 7 Neuroimaging Research Group, King's College London, PO42 De Crespigny Park, London, SE5 8AF, UK
| | - Fred H Gage
- 8 The Salk Institute for Biological Studies, 10010 N. Torrey Pines Road, La Jolla, CA 92037, USA
| | - Lawrence D F Moon
- 1 Neurorestoration Group, Wolfson Centre for Age-Related Diseases, King's College London, 16-18 Newcomen Street, London SE1 1UL, UK 2 Centre for Integrative Biology, King's College London, Franklin-Wilkins Building, 150 Stamford Street, London SE1 9NH, UK
| |
Collapse
|
19
|
Hang P, Zhao J, Cai B, Tian S, Huang W, Guo J, Sun C, Li Y, Du Z. Brain-derived neurotrophic factor regulates TRPC3/6 channels and protects against myocardial infarction in rodents. Int J Biol Sci 2015; 11:536-45. [PMID: 25892961 PMCID: PMC4400385 DOI: 10.7150/ijbs.10754] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2014] [Accepted: 02/23/2015] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Brain-derived neurotrophic factor (BDNF) is associated with coronary artery diseases. However, its role and mechanism in myocardial infarction (MI) is not fully understood. METHODS Wistar rat and Kunming mouse model of MI were induced by the ligation of left coronary artery. Blood samples were collected from MI rats and patients. Plasma BDNF level, protein expression of BDNF, tropomyosin-related kinase B (TrkB) and its downstream transient receptor potential canonical (TRPC)3/6 channels were examined by enzyme-linked immunosorbent assay and Western blot. Infarct size, cardiac function and cardiomyocyte apoptosis were measured after intra-myocardium injection with recombinant human BDNF. Protective role of BDNF against cardiomyocyte apoptosis was confirmed by BDNF scavenger TrkB-Fc. The regulation of TRPC3/6 channels by BDNF was validated by pretreating with TRPC blocker (2-Aminoethyl diphenylborinate, 2-APB) and TRPC3/6 siRNAs. RESULTS Circulating BDNF was significantly enhanced in MI rats and patients. Protein expression of BDNF, TrkB and TRPC3/6 channels were upregulated in MI. 3 days post-MI, BDNF treatment markedly reduced the infarct size and serum lactate dehydrogenase activity. Meanwhile, echocardiography indicated that BDNF significantly improved cardiac function of MI mice. Furthermore, BDNF markedly inhibited cardiomyocyte apoptosis by upregulating Bcl-2 expression and downregulating caspase-3 expression and activity in ischemic myocardium. In neonatal rat ventricular myocytes, cell viability was dramatically increased by BDNF in hypoxia, which was restored by TrkB-Fc. Furthermore, protective role of BDNF against hypoxia-induced apoptosis was reversed by 2-APB and TRPC3/6 siRNAs. CONCLUSION BDNF/TrkB alleviated cardiac ischemic injury and inhibited cardiomyocytes apoptosis by regulating TRPC3/6 channels, which provides a novel potential therapeutic candidate for MI.
Collapse
Affiliation(s)
- Pengzhou Hang
- 1. Institute of Clinical Pharmacology of the Second Affiliated Hospital (Key Laboratory of Drug Research, Heilongjiang Higher Education Institutions), Harbin Medical University, Harbin 150086, China
| | - Jing Zhao
- 2. Department of Cardiology of the First Affiliated Hospital (Key Laboratory of Cardiac Diseases and Heart Failure), Harbin Medical University, Harbin 150001, China
| | - Benzhi Cai
- 3. Department of Pharmacology (State-Province Key Laboratories of Biomedicine- Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), Harbin Medical University, Harbin 150081, China
| | - Shanshan Tian
- 1. Institute of Clinical Pharmacology of the Second Affiliated Hospital (Key Laboratory of Drug Research, Heilongjiang Higher Education Institutions), Harbin Medical University, Harbin 150086, China
| | - Wei Huang
- 1. Institute of Clinical Pharmacology of the Second Affiliated Hospital (Key Laboratory of Drug Research, Heilongjiang Higher Education Institutions), Harbin Medical University, Harbin 150086, China
| | - Jing Guo
- 1. Institute of Clinical Pharmacology of the Second Affiliated Hospital (Key Laboratory of Drug Research, Heilongjiang Higher Education Institutions), Harbin Medical University, Harbin 150086, China
| | - Chuan Sun
- 1. Institute of Clinical Pharmacology of the Second Affiliated Hospital (Key Laboratory of Drug Research, Heilongjiang Higher Education Institutions), Harbin Medical University, Harbin 150086, China
| | - Yue Li
- 2. Department of Cardiology of the First Affiliated Hospital (Key Laboratory of Cardiac Diseases and Heart Failure), Harbin Medical University, Harbin 150001, China
| | - Zhimin Du
- 1. Institute of Clinical Pharmacology of the Second Affiliated Hospital (Key Laboratory of Drug Research, Heilongjiang Higher Education Institutions), Harbin Medical University, Harbin 150086, China
| |
Collapse
|
20
|
Chen Q, Zhang Z, Liu J, He Q, Zhou Y, Shao G, Sun X, Cao X, Gong A, Jiang P. A fibrin matrix promotes the differentiation of EMSCs isolated from nasal respiratory mucosa to myelinating phenotypical Schwann-like cells. Mol Cells 2015; 38:221-8. [PMID: 25666351 PMCID: PMC4363721 DOI: 10.14348/molcells.2015.2170] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2014] [Revised: 10/08/2014] [Accepted: 11/19/2014] [Indexed: 12/16/2022] Open
Abstract
Because Schwann cells perform the triple tasks of myelination, axon guidance and neurotrophin synthesis, they are candidates for cell transplantation that might cure some types of nervous-system degenerative diseases or injuries. However, Schwann cells are difficult to obtain. As another option, ectomesenchymal stem cells (EMSCs) can be easily harvested from the nasal respiratory mucosa. Whether fibrin, an important transplantation vehicle, can improve the differentiation of EMSCs into Schwann-like cells (SLCs) deserves further research. EMSCs were isolated from rat nasal respiratory mucosa and were purified using anti-CD133 magnetic cell sorting. The purified cells strongly expressed HNK-1, nestin, p75(NTR), S-100, and vimentin. Using nuclear staining, the MTT assay and Western blotting analysis of the expression of cell-cycle markers, the proliferation rate of EMSCs on a fibrin matrix was found to be significantly higher than that of cells grown on a plastic surface but insignificantly lower than that of cells grown on fibronectin. Additionally, the EMSCs grown on the fibrin matrix expressed myelination-related molecules, including myelin basic protein (MBP), 2',3'-cyclic nucleotide 3'-phosphodiesterase (CNPase) and galactocerebrosides (GalCer), more strongly than did those grown on fibronectin or a plastic surface. Furthermore, the EMSCs grown on the fibrin matrix synthesized more neurotrophins compared with those grown on fibronectin or a plastic surface. The expression level of integrin in EMSCs grown on fibrin was similar to that of cells grown on fibronectin but was higher than that of cells grown on a plastic surface. These results demonstrated that fibrin not only promoted EMSC proliferation but also the differentiation of EMSCs into the SLCs. Our findings suggested that fibrin has great promise as a cell transplantation vehicle for the treatment of some types of nervous system diseases or injuries.
Collapse
Affiliation(s)
- Qian Chen
- Department of Histology and Embryology, School of Medicine, Jiangsu University, Zhenjiang,
China
| | - Zhijian Zhang
- Department of Histology and Embryology, School of Medicine, Jiangsu University, Zhenjiang,
China
| | - Jinbo Liu
- Department of Orthopedics, the Third Affiliated Hospital of Suzhou University, Changzhou,
China
| | - Qinghua He
- School of Pharmacology, Jiangsu University, Zhenjiang,
China
| | - Yuepeng Zhou
- Department of Histology and Embryology, School of Medicine, Jiangsu University, Zhenjiang,
China
| | - Genbao Shao
- Department of Histology and Embryology, School of Medicine, Jiangsu University, Zhenjiang,
China
| | - Xianglan Sun
- Department of Histology and Embryology, School of Medicine, Jiangsu University, Zhenjiang,
China
| | - Xudong Cao
- Department of Chemical Engineering, University of Ottawa, Ottawa, Ontario,
Canada
| | - Aihua Gong
- Department of Histology and Embryology, School of Medicine, Jiangsu University, Zhenjiang,
China
| | - Ping Jiang
- Department of Histology and Embryology, School of Medicine, Jiangsu University, Zhenjiang,
China
| |
Collapse
|
21
|
Abstract
Stroke is a leading cause of morbidity in the developed world and results in chronic disability in many cases. The literature related to the critical factors that regulate tissue self-regeneration in stroke is still limited, which restricts effective therapy. However, optimism in this area has been provided by recent research. The mechanisms involved in tissue regeneration and the mode of the participation of stem/progenitor cells and soluble protein neurotrophic factors in this process may yield a more complete understanding of the nature of stroke. This review summarizes the current understanding of both cellular and humoral issues with a particular emphasis on how these issues contribute to tissue regeneration in stroke.
Collapse
Affiliation(s)
- Bogusław Machalinski
- Department of General Pathology, Pomeranian Medical University, Al. Powstancow Wlkp. 72, Szczecin 70-111, Poland
| |
Collapse
|
22
|
Schizas N, Andersson B, Hilborn J, Hailer NP. Interleukin-1 receptor antagonist promotes survival of ventral horn neurons and suppresses microglial activation in mouse spinal cord slice cultures. J Neurosci Res 2014; 92:1457-65. [PMID: 24975034 DOI: 10.1002/jnr.23429] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2013] [Revised: 04/23/2014] [Accepted: 05/09/2014] [Indexed: 02/05/2023]
Abstract
Secondary damage after spinal cord injury (SCI) induces neuronal demise through neurotoxicity and inflammation, and interleukin (IL)-1β is a key inflammatory mediator. We hypothesized that IL-1β is released in spinal cord slice cultures (SCSC) and aimed at preventing the potentially neurotoxic effects of IL-1β by using interleukin-1 receptor antagonist (IL1RA). We hypothesized that IL1RA treatment enhances neuronal survival and suppresses microglial activation. SCSC were cultured up to 8 days in vitro (DIV) in the presence of IL1RA or without, either combined with trophic support using neurotrophin (NT)-3 or not. Four groups were studied: negative control, IL1RA, NT-3, and IL1RA + NT-3. IL-1β concentrations in supernatants were measured by ELISA. SCSC were immunohistochemically stained for NeuN and α-neurofilament, and microglial cells were visualized with isolectin B4 . After 8 DIV, ventral horn neurons were significantly more numerous in the IL1RA, NT-3, and IL1RA + NT-3 groups compared with negative controls. Activated microglial cells were significantly less numerous in the IL1RA, NT-3, and IL1RA + NT-3 groups compared with negative controls. Axons expanded into the collagen matrix after treatment with IL1RA, NT-3, or IL1RA + NT-3, but not in negative controls. IL-1β release from cultures peaked after 6 hr and was lowest in the IL1RA + NT-3 group. We conclude that IL-1β is released in traumatized spinal cord tissue and that IL1RA could exert its neuroprotective actions by blocking IL-1-receptors. IL1RA thereby sustains neuronal survival irrespective of the presence of additional trophic support. Microglial activation is suppressed in the presence of IL1RA, suggesting decreased inflammatory activity. IL1RA treatment approaches may have substantial impact following SCI.
Collapse
Affiliation(s)
- N Schizas
- The SpineLab, Institute of Surgical Sciences, Department of Orthopaedics, Uppsala University, SE-751 85, Uppsala, Sweden
| | | | | | | |
Collapse
|
23
|
Shen W, Zhu L, Lee SR, Chung SH, Gillies MC. Involvement of NT3 and P75(NTR) in photoreceptor degeneration following selective Müller cell ablation. J Neuroinflammation 2013; 10:137. [PMID: 24224958 PMCID: PMC3831588 DOI: 10.1186/1742-2094-10-137] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2013] [Accepted: 11/01/2013] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Neurotrophins can regulate opposing functions that result in cell survival or apoptosis, depending on which form of the protein is secreted and which receptor and signaling pathway is activated. We have recently developed a transgenic model in which inducible and patchy Müller cell ablation leads to photoreceptor degeneration. This study aimed to examine the roles of mature neurotrophin-3 (NT3), pro-NT3 and p75 neurotrophin receptor (P75(NTR)) in photoreceptor degeneration in this model. METHODS Transgenic mice received tamoxifen to induce Müller cell ablation. Changes in the status of Müller and microglia cells as well as expression of mature NT3, pro-NT3 and P75(NTR) were examined by immunohistochemistry and Western blot analysis. Recombinant mature NT3 and an antibody neutralizing 75(NTR) were injected intravitreally 3 and 6 days after Müller cell ablation to examine their effects on photoreceptor degeneration and microglial activation. RESULTS We found that patchy loss of Müller cells was associated with activation of surviving Müller cells and microglial cells, concurrently with reduced expression of mature NT3 and upregulation of pro-NT3 and P75(NTR). Intravitreal injection of mature NT3 and a neutralizing antibody to P75NTR, either alone or in combination, attenuated photoreceptor degeneration and the beneficial effect was associated with inhibition of microglial activation. CONCLUSIONS Our data suggest that Müller cell ablation alters the balance between the protective and deleterious effects of mature NT3 and pro-NT3. Modulation of the neuroprotective action of mature NT3 and pro-apoptotic pro-NT3/P75(NTR) signaling may represent a novel pharmacological strategy for photoreceptor protection in retinal disease.
Collapse
Affiliation(s)
- Weiyong Shen
- Save Sight Institute, the University of Sydney, 8 Macquarie Street, Sydney 2000, Australia
| | - Ling Zhu
- Save Sight Institute, the University of Sydney, 8 Macquarie Street, Sydney 2000, Australia
| | - So-Ra Lee
- Save Sight Institute, the University of Sydney, 8 Macquarie Street, Sydney 2000, Australia
| | - Sook H Chung
- Save Sight Institute, the University of Sydney, 8 Macquarie Street, Sydney 2000, Australia
| | - Mark C Gillies
- Save Sight Institute, the University of Sydney, 8 Macquarie Street, Sydney 2000, Australia
| |
Collapse
|
24
|
D'Onofrio PM, Koeberle PD. What can we learn about stroke from retinal ischemia models? Acta Pharmacol Sin 2013. [PMID: 23202803 DOI: 10.1038/aps.2012.165] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Retinal ischemia is a very useful model to study the impact of various cell death pathways, such as apoptosis and necrosis, in the ischemic retina. However, it is important to note that the retina is formed as an outpouching of the diencephalon and is part of the central nervous system. As such, the cell death pathways initiated in response to ischemic damage in the retina reflect those found in other areas of the central nervous system undergoing similar trauma. The retina is also more accessible than other areas of the central nervous system, thus making it a simpler model to work with and study. By utilizing the retinal model, we can greatly increase our knowledge of the cell death processes initiated by ischemia which lead to degeneration in the central nervous system. This paper examines work that has been done so far to characterize various aspects of cell death in the retinal ischemia model, such as various pathways which are activated, and the role neurotrophic factors, and discusses how these are relevant to the treatment of ischemic damage in both the retina and the greater central nervous system.
Collapse
|