Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.426
Filter
1.
Int J Nanomedicine ; 19: 4081-4101, 2024.
Article in English | MEDLINE | ID: mdl-38736654

ABSTRACT

Purpose: Spinal cord injury (SCI) is an incurable and disabling event that is accompanied by complex inflammation-related pathological processes, such as the production of excessive reactive oxygen species (ROS) by infiltrating inflammatory immune cells and their release into the extracellular microenvironment, resulting in extensive apoptosis of endogenous neural stem cells. In this study, we noticed the neuroregeneration-promoting effect as well as the ability of the innovative treatment method of FTY720-CDs@GelMA paired with NSCs to increase motor function recovery in a rat spinal cord injury model. Methods: Carbon dots (CDs) and fingolimod (FTY720) were added to a hydrogel created by chemical cross-linking GelMA (FTY720-CDs@GelMA). The basic properties of FTY720-CDs@GelMA hydrogels were investigated using TEM, SEM, XPS, and FTIR. The swelling and degradation rates of FTY720-CDs@GelMA hydrogels were measured, and each group's ability to scavenge reactive oxygen species was investigated. The in vitro biocompatibility of FTY720-CDs@GelMA hydrogels was assessed using neural stem cells. The regeneration of the spinal cord and recovery of motor function in rats were studied following co-treatment of spinal cord injury using FTY720-CDs@GelMA hydrogel in combination with NSCs, utilising rats with spinal cord injuries as a model. Histological and immunofluorescence labelling were used to determine the regeneration of axons and neurons. The recovery of motor function in rats was assessed using the BBB score. Results: The hydrogel boosted neurogenesis and axonal regeneration by eliminating excess ROS and restoring the regenerative environment. The hydrogel efficiently contained brain stem cells and demonstrated strong neuroprotective effects in vivo by lowering endogenous ROS generation and mitigating ROS-mediated oxidative stress. In a follow-up investigation, we discovered that FTY720-CDs@GelMA hydrogel could dramatically boost NSC proliferation while also promoting neuronal regeneration and synaptic formation, hence lowering cavity area. Conclusion: Our findings suggest that the innovative treatment of FTY720-CDs@GelMA paired with NSCs can effectively improve functional recovery in SCI patients, making it a promising therapeutic alternative for SCI.


Subject(s)
Fingolimod Hydrochloride , Hydrogels , Neural Stem Cells , Rats, Sprague-Dawley , Spinal Cord Injuries , Animals , Spinal Cord Injuries/drug therapy , Spinal Cord Injuries/therapy , Fingolimod Hydrochloride/pharmacology , Fingolimod Hydrochloride/chemistry , Fingolimod Hydrochloride/administration & dosage , Neural Stem Cells/drug effects , Hydrogels/chemistry , Hydrogels/pharmacology , Hydrogels/administration & dosage , Rats , Recovery of Function/drug effects , Reactive Oxygen Species/metabolism , Quantum Dots/chemistry , Disease Models, Animal , Female , Spinal Cord/drug effects
2.
Int J Nanomedicine ; 19: 4181-4197, 2024.
Article in English | MEDLINE | ID: mdl-38766656

ABSTRACT

Purpose: The committed differentiation fate regulation has been a difficult problem in the fields of stem cell research, evidence showed that nanomaterials could promote the differentiation of stem cells into specific cell types. Layered double hydroxide (LDH) nanoparticles possess the regulation function of stem cell fate, while the underlying mechanism needs to be investigated. In this study, the process of embryonic stem cells (ESCs) differentiate to neural progenitor cells (NPCs) by magnesium aluminum LDH (MgAl-LDH) was investigated. Methods: MgAl-LDH with diameters of 30, 50, and 100 nm were synthesized and characterized, and their effects on the cytotoxicity and differentiation of NPCs were detected in vitro. Dot blot and MeRIP-qPCR were performed to detect the level of m6A RNA methylation in nanoparticles-treated cells. Results: Our work displayed that LDH nanoparticles of three different sizes were biocompatible with NPCs, and the addition of MgAl-LDH could significantly promote the process of ESCs differentiate to NPCs. 100 nm LDH has a stronger effect on promoting NPCs differentiation compared to 30 nm and 50 nm LDH. In addition, dot blot results indicated that the enhanced NPCs differentiation by MgAl-LDH was closely related to m6A RNA methylation process, and the major modification enzyme in LDH controlled NPCs differentiation may be the m6A RNA methyltransferase METTL3. The upregulated METTL3 by LDH increased the m6A level of Sox1 mRNA, enhancing its stability. Conclusion: This work reveals that MgAl-LDH nanoparticles can regulate the differentiation of ESCs into NPCs by increasing m6A RNA methylation modification of Sox1.


Subject(s)
Cell Differentiation , Nanoparticles , Neural Stem Cells , Cell Differentiation/drug effects , Animals , Neural Stem Cells/drug effects , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Mice , Nanoparticles/chemistry , Methylation/drug effects , Hydroxides/chemistry , Hydroxides/pharmacology , Methyltransferases/metabolism , Methyltransferases/genetics , Particle Size , Embryonic Stem Cells/drug effects , Embryonic Stem Cells/cytology , Adenosine/pharmacology , Adenosine/chemistry , Adenosine/analogs & derivatives , Aluminum Hydroxide/chemistry , Aluminum Hydroxide/pharmacology , Magnesium Hydroxide/chemistry , Magnesium Hydroxide/pharmacology
3.
Neuron ; 112(9): 1373-1375, 2024 May 01.
Article in English | MEDLINE | ID: mdl-38697018

ABSTRACT

Maternal well-being is important for the development of the fetus, with a key influence on its nervous system. In this issue of Neuron, Krontira et al.1 implicate glucocorticoids, the stress hormones, in the regulation of neural stem cell identity and proliferation, with long-lasting consequences on brain architecture and educational attainment.


Subject(s)
Glucocorticoids , Neurogenesis , Humans , Glucocorticoids/pharmacology , Neurogenesis/drug effects , Neurogenesis/physiology , Neurons/drug effects , Neurons/physiology , Cerebral Cortex/drug effects , Cerebral Cortex/cytology , Neural Stem Cells/drug effects
4.
Zhongguo Zhong Yao Za Zhi ; 49(9): 2316-2325, 2024 May.
Article in Chinese | MEDLINE | ID: mdl-38812132

ABSTRACT

This study aimed to investigate the intervention effect of tetramethylpyrazine(TMP) combined with transplantation of neural stem cells(NSCs) on middle cerebral artery occlusion(MCAO) rat model and to explore the mechanism of TMP combined with NSCs transplantation on ischemic stroke based on the regulation of stem cell biological behavior. MCAO rats were randomly divided into a model group, a TMP group, an NSCs transplantation group, and a TMP combined with NSCs transplantation group according to neurological function scores. A sham group was set up at the same time. The neurological function score was used to evaluate the improvement of neurological function in MCAO rats after TMP combined with NSCs transplantation. The proliferation, migration, and differentiation of NSCs were evaluated by BrdU, BrdU/DCX, BrdU/NeuN, and BrdU/GFAP immunofluorescence labeling. The protein expression of stromal cell-derived factor 1(SDF-1), C-X-C motif chemokine receptor 4(CXCR4), as well as oxidative stress pathway proteins nuclear factor erythroid 2-related factor 2(Nrf2), Kelch-like ECH-associated protein 1(KEAP1), heme oxygenase 1(HO-1), NAD(P)H quinone oxidoreductase 1(NQO1) was detected by Western blot to study the migration mechanism of TMP combined with NSCs. The results showed that TMP combined with NSCs transplantation significantly improved the neurological function score in MCAO rats. Immunofluorescence staining showed a significant increase in the number of BrdU~+, BrdU~+/DCX~+, BrdU~+/NeuN~+, and BrdU~+/GFAP~+ cells in the TMP, NSCs transplantation, and combined treatment groups, with the combined treatment group showing the most significant increase. Further Western blot analysis revealed significantly elevated expression of CXCR4 protein in the TMP, NSCs transplantation, and combined treatment groups, along with up-regulated protein expression of Nrf2, HO-1, and NQO1, and decreased KEAP1 protein expression. This study showed that both TMP and NSCs transplantation can promote the recovery of neurological function by promoting the proliferation, migration, and differentiation of NSCs, and the effect of TMP combined with NSCs transplantation is superior. The mechanism of action may be related to the activation of the Nrf2/HO-1/CXCR4 pathway.


Subject(s)
Brain Ischemia , Doublecortin Protein , NF-E2-Related Factor 2 , Neural Stem Cells , Pyrazines , Rats, Sprague-Dawley , Receptors, CXCR4 , Animals , Pyrazines/pharmacology , Neural Stem Cells/drug effects , Neural Stem Cells/transplantation , Neural Stem Cells/metabolism , Rats , Male , Receptors, CXCR4/metabolism , Receptors, CXCR4/genetics , Brain Ischemia/therapy , Brain Ischemia/metabolism , Brain Ischemia/drug therapy , NF-E2-Related Factor 2/metabolism , NF-E2-Related Factor 2/genetics , Chemokine CXCL12/metabolism , Chemokine CXCL12/genetics , Kelch-Like ECH-Associated Protein 1/metabolism , Kelch-Like ECH-Associated Protein 1/genetics , Stem Cell Transplantation/methods , Cell Proliferation/drug effects , Cell Movement/drug effects , Humans , Reperfusion Injury/therapy , Reperfusion Injury/metabolism , Infarction, Middle Cerebral Artery/therapy , NAD(P)H Dehydrogenase (Quinone)/metabolism , NAD(P)H Dehydrogenase (Quinone)/genetics
5.
Zhongguo Zhong Yao Za Zhi ; 49(9): 2308-2315, 2024 May.
Article in Chinese | MEDLINE | ID: mdl-38812131

ABSTRACT

This study aims to decipher the mechanism of tetramethylpyrazine(TMP) in regulating the migration of neural stem cells(NSCs) in the rat model of middle cerebral artery occlusion(MCAO) via the nuclear factor erythroid 2-related factor 2(Nrf2)/heme oxygenase 1(HO-1)/C-X-C motif chemokine receptor 4(CXCR4) pathway. SD rats were randomized into sham, MCAO(model), and tetramethylpyrazine(TMP, 20 mg·kg~(-1) and 40 mg·kg~(-1)) groups. The neurological impairment was assessed by the modified neurological severity score(mNSS). The immunofluorescence assay was employed to detect the cells stained with both 5-bromodeoxyuridine(BrdU) and doublecortin(DCX) in the brain tissue. The effect of TMP on the migration of C17.2 cells was observed. Western blot was employed to determine the protein levels of Nrf2, HO-1, p62, NAD(P)H quinone oxidoreductase 1(NQO1), stromal cell-derived factor 1(SDF-1), and CXCR4 in the brain tissue and C17.2 cells. The results showed that after 7 days and 21 days of mode-ling, the mNSS and BrdU~+/DCX~+ cells were increased, and the expression of Nrf2 and CXCR4 in the brain tissue was up-regulated. Compared with the model group, TMP(40 mg·kg~(-1)) reduced the mNSS, increased the number of BrdU~+/DCX~+ cells, and up-regulated the expression of Nrf2, CXCR4, and SDF-1. In addition, TMP promoted the migration of C17.2 cells and up-regulated the expression of p62, Nrf2, HO-1, and NQO1 in a time-and dose-dependent manner. The expression was the highest at the time point of 12 h in the TMP(50 µg·mL~(-1)) group(P<0.01). In conclusion, TMP activates the Nrf2/HO-1/CXCR4 pathway to promote the migration of NSCs to the ischemic area, thus exerting the therapeutic effect on the ischemia-reperfusion injury. This study provides experimental support for the application of TMP in ischemic stroke.


Subject(s)
Cell Movement , Heme Oxygenase-1 , NF-E2-Related Factor 2 , Neural Stem Cells , Pyrazines , Rats, Sprague-Dawley , Receptors, CXCR4 , Animals , Receptors, CXCR4/metabolism , Receptors, CXCR4/genetics , NF-E2-Related Factor 2/metabolism , NF-E2-Related Factor 2/genetics , Pyrazines/pharmacology , Rats , Neural Stem Cells/drug effects , Neural Stem Cells/metabolism , Neural Stem Cells/cytology , Cell Movement/drug effects , Male , Heme Oxygenase-1/metabolism , Heme Oxygenase-1/genetics , Doublecortin Protein , Signal Transduction/drug effects , Reperfusion Injury/metabolism , Reperfusion Injury/drug therapy , Humans
6.
Biosensors (Basel) ; 14(5)2024 Apr 23.
Article in English | MEDLINE | ID: mdl-38785686

ABSTRACT

Combinatorial drug therapy has emerged as a critically important strategy in medical research and patient treatment and involves the use of multiple drugs in concert to achieve a synergistic effect. This approach can enhance therapeutic efficacy while simultaneously mitigating adverse side effects. However, the process of identifying optimal drug combinations, including their compositions and dosages, is often a complex, costly, and time-intensive endeavor. To surmount these hurdles, we propose a novel microfluidic device capable of simultaneously generating multiple drug concentration gradients across an interlinked array of culture chambers. This innovative setup allows for the real-time monitoring of live cell responses. With minimal effort, researchers can now explore the concentration-dependent effects of single-agent and combination drug therapies. Taking neural stem cells (NSCs) as a case study, we examined the impacts of various growth factors-epithelial growth factor (EGF), platelet-derived growth factor (PDGF), and fibroblast growth factor (FGF)-on the differentiation of NSCs. Our findings indicate that an overdose of any single growth factor leads to an upsurge in the proportion of differentiated NSCs. Interestingly, the regulatory effects of these growth factors can be modulated by the introduction of additional growth factors, whether singly or in combination. Notably, a reduced concentration of these additional factors resulted in a decreased number of differentiated NSCs. Our results affirm that the successful application of this microfluidic device for the generation of multi-drug concentration gradients has substantial potential to revolutionize drug combination screening. This advancement promises to streamline the process and accelerate the discovery of effective therapeutic drug combinations.


Subject(s)
High-Throughput Screening Assays , Neural Stem Cells , Neural Stem Cells/drug effects , Humans , Cell Differentiation , Lab-On-A-Chip Devices , Platelet-Derived Growth Factor , Epidermal Growth Factor , Drug Evaluation, Preclinical , Drug Combinations , Fibroblast Growth Factors
7.
Sci Rep ; 14(1): 12167, 2024 05 28.
Article in English | MEDLINE | ID: mdl-38806485

ABSTRACT

During neuroinflammation, monocytes that infiltrate the central nervous system (CNS) may contribute to regenerative processes depending on their activation status. However, the extent and mechanisms of monocyte-induced CNS repair in patients with neuroinflammatory diseases remain largely unknown, partly due to the lack of a fully human assay platform that can recapitulate monocyte-neural stem cell interactions within the CNS microenvironment. We therefore developed a human model system to assess the impact of monocytic factors on neural stem cells, establishing a high-content compatible assay for screening monocyte-induced neural stem cell proliferation and differentiation. The model combined monocytes isolated from healthy donors and human embryonic stem cell derived neural stem cells and integrated both cell-intrinsic and -extrinsic properties. We identified CNS-mimicking culture media options that induced a monocytic phenotype resembling CNS infiltrating monocytes, while allowing adequate monocyte survival. Monocyte-induced proliferation, gliogenic fate and neurogenic fate of neural stem cells were affected by the conditions of monocytic priming and basal neural stem cell culture as extrinsic factors as well as the neural stem cell passage number as an intrinsic neural stem cell property. We developed a high-content compatible human in vitro assay for the integrated analysis of monocyte-derived factors on CNS repair.


Subject(s)
Cell Differentiation , Cell Proliferation , Monocytes , Neural Stem Cells , Humans , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Neural Stem Cells/drug effects , Monocytes/cytology , Monocytes/metabolism , Monocytes/drug effects , Cell Proliferation/drug effects , Cell Differentiation/drug effects , Cells, Cultured
8.
Bull Exp Biol Med ; 176(5): 576-580, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38724808

ABSTRACT

We performed a comparative in vitro study of the involvement of NF-κB, PI3K, cAMP, ERK1/2, p38, JAKs, STAT3, JNK, and p53-dependent intracellular signaling in the functioning of neural stem cells (NSC) under the influence of basic fibroblast growth factor (FGF) and FGF receptor agonist, diterpene alkaloid songorine. The significant differences in FGFR-mediated intracellular signaling in NSC were revealed for these ligands. In both cases, stimulation of progenitor cell proliferation occurs with the participation of NF-κB, PI3K, ERK1/2, JAKs, and STAT3, while JNK and p53, on the contrary, inhibit cell cycle progression. However, under the influence of songorin, cAMP- and p38-mediated cascades are additionally involved in the transmission of the NSC division-activating signal. In addition, unlike FGF, the alkaloid stimulates progenitor cell differentiation by activating ERK1/2, p38, JNK, p53, and STAT3.


Subject(s)
Cell Differentiation , Cell Proliferation , Diterpenes , Neural Stem Cells , Receptors, Fibroblast Growth Factor , STAT3 Transcription Factor , Signal Transduction , Neural Stem Cells/drug effects , Neural Stem Cells/metabolism , Neural Stem Cells/cytology , Animals , STAT3 Transcription Factor/metabolism , Receptors, Fibroblast Growth Factor/metabolism , Receptors, Fibroblast Growth Factor/agonists , Signal Transduction/drug effects , Cell Proliferation/drug effects , Diterpenes/pharmacology , Cell Differentiation/drug effects , NF-kappa B/metabolism , Fibroblast Growth Factor 2/metabolism , Fibroblast Growth Factor 2/pharmacology , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Protein p53/agonists , Phosphatidylinositol 3-Kinases/metabolism , Alkaloids/pharmacology , MAP Kinase Signaling System/drug effects , Janus Kinases/metabolism , Cyclic AMP/metabolism , Cells, Cultured , Rats
9.
Biomater Adv ; 161: 213895, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38795474

ABSTRACT

Ischemic stroke, a cerebrovascular disease caused by arterial occlusion in the brain, can lead to brain impairment and even death. Stem cell therapies have shown positive advantages to treat ischemic stroke because of their extended time window, but the cell viability is poor when transplanted into the brain directly. Therefore, a new hydrogel GelMA-T was developed by introducing taurine on GelMA to transplant neural stem cells. The GelMA-T displayed the desired photocuring ability, micropore structure, and cytocompatibility. Its compressive modulus was more similar to neural tissue compared to that of GelMA. The GelMA-T could protect SH-SY5Y cells from injury induced by OGD/R. Furthermore, the NE-4C cells showed better proliferation performance in GelMA-T than that in GelMA during both 2D and 3D cultures. All results demonstrate that GelMA-T possesses a neuroprotective effect for ischemia/reperfusion injury against ischemic stroke and plays a positive role in promoting NSC proliferation. The novel hydrogel is anticipated to function as cell vehicles for the transplantation of neural stem cells into the stroke cavity, aiming to treat ischemic stroke.


Subject(s)
Cell Proliferation , Hydrogels , Neural Stem Cells , Neuroprotective Agents , Taurine , Neural Stem Cells/drug effects , Neural Stem Cells/transplantation , Taurine/pharmacology , Cell Proliferation/drug effects , Neuroprotective Agents/pharmacology , Hydrogels/chemistry , Hydrogels/pharmacology , Humans , Animals , Cell Survival/drug effects
10.
Biosci Rep ; 44(5)2024 May 29.
Article in English | MEDLINE | ID: mdl-38700092

ABSTRACT

Pre-eclampsia (PE) is a hypertensive disorder of pregnancy which is associated with increased risk of neurodevelopmental disorders in exposed offspring. The pathophysiological mechanisms mediating this relationship are currently unknown, and one potential candidate is the anti-angiogenic factor soluble Fms-like tyrosine kinase 1 (sFlt-1), which is highly elevated in PE. While sFlt-1 can impair angiogenesis via inhibition of VEGFA signalling, it is unclear whether it can directly affect neuronal development independently of its effects on the vasculature. To test this hypothesis, the current study differentiated the human neural progenitor cell (NPC) line ReNcell® VM into a mixed culture of mature neurons and glia, and exposed them to sFlt-1 during development. Outcomes measured were neurite growth, cytotoxicity, mRNA expression of nestin, MBP, GFAP, and ßIII-tubulin, and neurosphere differentiation. sFlt-1 induced a significant reduction in neurite growth and this effect was timing- and dose-dependent up to 100 ng/ml, with no effect on cytotoxicity. sFlt-1 (100 ng/ml) also reduced ßIII-tubulin mRNA and neuronal differentiation of neurospheres. Undifferentiated NPCs and mature neurons/glia expressed VEGFA and VEGFR-2, required for endogenous autocrine and paracrine VEGFA signalling, while sFlt-1 treatment prevented the neurogenic effects of exogenous VEGFA. Overall, these data provide the first experimental evidence for a direct effect of sFlt-1 on neurite growth and neuronal differentiation in human neurons through inhibition of VEGFA signalling, clarifying our understanding of the potential role of sFlt-1 as a mechanism by which PE can affect neuronal development.


Subject(s)
Cell Differentiation , Neural Stem Cells , Neurons , Vascular Endothelial Growth Factor Receptor-1 , Humans , Vascular Endothelial Growth Factor Receptor-1/metabolism , Vascular Endothelial Growth Factor Receptor-1/genetics , Neural Stem Cells/metabolism , Neural Stem Cells/drug effects , Neurons/metabolism , Neurons/drug effects , Neurons/cytology , Cell Differentiation/drug effects , Neurites/metabolism , Neurites/drug effects , Neurogenesis/drug effects , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor A/genetics , Female , Pre-Eclampsia/metabolism , Pre-Eclampsia/pathology , Pregnancy , Cell Line, Tumor , Signal Transduction
11.
eNeuro ; 11(6)2024 Jun.
Article in English | MEDLINE | ID: mdl-38729764

ABSTRACT

Intracerebral hemorrhage (ICH), the most common subtype of hemorrhagic stroke, leads to cognitive impairment and imposes significant psychological burdens on patients. Hippocampal neurogenesis has been shown to play an essential role in cognitive function. Our previous study has shown that tetrahydrofolate (THF) promotes the proliferation of neural stem cells (NSCs). However, the effect of THF on cognition after ICH and the underlying mechanisms remain unclear. Here, we demonstrated that administration of THF could restore cognition after ICH. Using Nestin-GFP mice, we further revealed that THF enhanced the proliferation of hippocampal NSCs and neurogenesis after ICH. Mechanistically, we found that THF could prevent ICH-induced elevated level of PTEN and decreased expressions of phosphorylated AKT and mTOR. Furthermore, conditional deletion of PTEN in NSCs of the hippocampus attenuated the inhibitory effect of ICH on the proliferation of NSCs and abnormal neurogenesis. Taken together, these results provide molecular insights into ICH-induced cognitive impairment and suggest translational clinical therapeutic strategy for hemorrhagic stroke.


Subject(s)
Cognitive Dysfunction , Hippocampus , Neural Stem Cells , Neurogenesis , PTEN Phosphohydrolase , Signal Transduction , Tetrahydrofolates , Animals , Neurogenesis/drug effects , Neurogenesis/physiology , Hippocampus/drug effects , Hippocampus/metabolism , Cognitive Dysfunction/drug therapy , Cognitive Dysfunction/etiology , Cognitive Dysfunction/metabolism , PTEN Phosphohydrolase/metabolism , Male , Signal Transduction/drug effects , Signal Transduction/physiology , Neural Stem Cells/drug effects , Neural Stem Cells/metabolism , Tetrahydrofolates/pharmacology , Mice , Hemorrhagic Stroke , Mice, Inbred C57BL , Mice, Transgenic , Cell Proliferation/drug effects
12.
Chemosphere ; 358: 142124, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38677614

ABSTRACT

Metformin, the most commonly prescribed drug for the treatment of diabetes, is increasingly used during pregnancy to address various disorders such as diabetes, obesity, preeclampsia, and metabolic diseases. However, its impact on neocortex development remains unclear. Here, we investigated the direct effects of metformin on neocortex development, focusing on ERK and p35/CDK5 regulation. Using a pregnant rat model, we found that metformin treatment during pregnancy induces small for gestational age (SGA) and reduces relative cortical thickness in embryos and neonates. Additionally, we discovered that metformin inhibits neural progenitor cell proliferation in the sub-ventricular zone (SVZ)/ventricular zone (VZ) of the developing neocortex, a process possibly mediated by ERK inactivation. Furthermore, metformin induces neuronal apoptosis in the SVZ/VZ area of the developing neocortex. Moreover, metformin retards neuronal migration, cortical lamination, and differentiation, potentially through p35/CDK5 inhibition in the developing neocortex. Remarkably, compensating for p35 through in utero electroporation partially rescues metformin-impaired neuronal migration and development. In summary, our study reveals that metformin disrupts neocortex development by inhibiting neuronal progenitor proliferation, neuronal migration, cortical layering, and cortical neuron maturation, likely via ERK and p35/CDK5 inhibition. Consequently, our findings advocate for caution in metformin usage during pregnancy, given its potential adverse effects on fetal brain development.


Subject(s)
Cell Proliferation , Cyclin-Dependent Kinase 5 , Metformin , Neocortex , Metformin/pharmacology , Animals , Female , Pregnancy , Neocortex/drug effects , Cyclin-Dependent Kinase 5/metabolism , Rats , Cell Proliferation/drug effects , Neural Stem Cells/drug effects , Neural Stem Cells/metabolism , MAP Kinase Signaling System/drug effects , Neurons/drug effects , Rats, Sprague-Dawley , Cell Differentiation/drug effects , Neurogenesis/drug effects , Cell Movement/drug effects , Apoptosis/drug effects , Signal Transduction/drug effects
13.
Food Chem Toxicol ; 188: 114684, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38663761

ABSTRACT

Exposure to mercury and its organic form methylmercury (MeHg), is of great concern for the developing nervous system. Despite available literature on MeHg neurotoxicity, there is still uncertainty about its mechanisms of action and the doses that trigger developmental effects. Our study combines two alternative methodologies, the human neural stem cells (NSC) and the zebrafish (ZF) embryo, to address the neurotoxic effects of early exposure to nanomolar concentrations of MeHg. Our results show linear or nonmonotonic (hormetic) responses depending on studied parameters. In ZF, we observed a hormetic response in locomotion and larval rotation, but a concentration-dependent response for sensory organ size and habituation. We also observed a possible delayed response as MeHg had greater effects on larval activity at 5 days than at 24 h. In NSC cells, some parameters show a clear dose dependence, such as increased apoptosis and differentiation to glial cells or decreased neuronal precursors; while others show a hormetic response: neuronal differentiation or cell proliferation. This study shows that the ZF model was more susceptible than NSC to MeHg neurotoxicity. The combination of different models has improved the understanding of the underlying mechanisms of toxicity and possible compensatory mechanisms at the cellular and organismal level.


Subject(s)
Embryo, Nonmammalian , Methylmercury Compounds , Neural Stem Cells , Zebrafish , Methylmercury Compounds/toxicity , Zebrafish/embryology , Animals , Neural Stem Cells/drug effects , Humans , Embryo, Nonmammalian/drug effects , Cell Differentiation/drug effects , Dose-Response Relationship, Drug , Apoptosis/drug effects , Cell Proliferation/drug effects
14.
Int J Biol Macromol ; 269(Pt 2): 131720, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38677692

ABSTRACT

The human nervous system is an incredibly intricate physiological network, and neural cells lack the ability to repair and regenerate after a brain injury. 3-dimensional (3D) bioprinting technology offers a promising strategy for constructing biomimetic organ constructs and in vitro brain/disease models. The bioink serves as a pivotal component that emulates the microenvironment of biomimetic construct and exerts a profound influence on cellular behaviors. In this study, a series of mechanically adjustable and dual crosslinking bioinks were developed using photocrosslinkable methacrylated silk fibroin (SilMA) in combination with the ionic crosslinking material, pectin, or pectin methacryloyl (PecMA) with silk fibroin (SF) supplementation. SilMA/pectin exhibited superior properties, with SilMA providing biocompatibility and adjustable mechanical properties, while the addition of pectin enhanced printability. The porous structure supported neural cell growth, and 15 % SilMA/0.5 % pectin bioinks displayed excellent printability and shape fidelity. Neural stem/progenitor cells (NSPCs)-loaded bioinks were used to construct a 3D brain model, demonstrating sustained vitality and high neuronal differentiation without the need for growth factors. The SilMA/pectin bioinks demonstrated adjustable mechanical properties, favorable biocompatibility, and an environment highly conducive to neural induction, offering an alternative approach for neural tissue engineering applications or in vitro brain models.


Subject(s)
Bioprinting , Fibroins , Neural Stem Cells , Pectins , Printing, Three-Dimensional , Spheroids, Cellular , Pectins/chemistry , Fibroins/chemistry , Neural Stem Cells/cytology , Neural Stem Cells/drug effects , Neural Stem Cells/metabolism , Bioprinting/methods , Spheroids, Cellular/cytology , Tissue Scaffolds/chemistry , Animals , Tissue Engineering/methods , Biocompatible Materials/chemistry , Biocompatible Materials/pharmacology , Humans , Cell Differentiation/drug effects , Ink
15.
Acta Biomater ; 180: 308-322, 2024 May.
Article in English | MEDLINE | ID: mdl-38615813

ABSTRACT

Motor functional improvement represents a paramount treatment objective in the post-spinal cord injury (SCI) recovery process. However, neuronal cell death and axonal degeneration following SCI disrupt neural signaling, impeding the motor functional recovery. In this study, we developed a multifunctional decellularized spinal cord-derived extracellular matrix (dSECM), crosslinked with glial cell-derived neurotrophic factor (GDNF), to promote differentiation of stem cells into neural-like cells and facilitate axonogenesis and remyelination. After decellularization, the immunogenic cellular components were effectively removed in dSECM, while the crucial protein components were retained which supports stem cells proliferation and differentiation. Furthermore, sustained release of GDNF from the dSECM facilitated axonogenesis and remyelination by activating the PI3K/Akt and MEK/Erk pathways. Our findings demonstrate that the dSECM-GDNF platform promotes neurogenesis, axonogenesis, and remyelination to enhance neural signaling, thereby yielding promising therapeutic effects for motor functional improvement after SCI. STATEMENT OF SIGNIFICANCE: The dSECM promotes the proliferation and differentiation of MSCs or NSCs by retaining proteins associated with positive regulation of neurogenesis and neuronal differentiation, while eliminating proteins related to negative regulation of neurogenesis. After crosslinking, GDNF can be gradually released from the platform, thereby promoting neural differentiation, axonogenesis, and remyelination to enhance neural signaling through activation of the PI3K/Akt and MEK/Erk pathways. In vivo experiments demonstrated that dSECM-GDNF/MSC@GelMA hydrogel exhibited the ability to facilitate neuronal regeneration at 4 weeks post-surgery, while promoting axonogenesis and remyelination at 8 weeks post-surgery, ultimately leading to enhanced motor functional recovery. This study elucidates the ability of neural regeneration strategy to promote motor functional recovery and provides a promising approach for designing multifunctional tissue for SCI treatment.


Subject(s)
Extracellular Matrix , Glial Cell Line-Derived Neurotrophic Factor , Neurogenesis , Remyelination , Spinal Cord Injuries , Animals , Female , Rats , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Extracellular Matrix/metabolism , Glial Cell Line-Derived Neurotrophic Factor/pharmacology , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Neural Stem Cells/drug effects , Neural Stem Cells/metabolism , Neural Stem Cells/cytology , Neurogenesis/drug effects , Rats, Sprague-Dawley , Recovery of Function/drug effects , Remyelination/drug effects , Spinal Cord Injuries/therapy , Spinal Cord Injuries/pathology
16.
Biofabrication ; 16(3)2024 May 09.
Article in English | MEDLINE | ID: mdl-38565133

ABSTRACT

Spinal cord injury (SCI) can cause permanent impairment to motor or sensory functions. Pre-cultured neural stem cell (NSC) hydrogel scaffolds have emerged as a promising approach to treat SCI by promoting anti-inflammatory effects, axon regrowth, and motor function restoration. Here, in this study, we performed a coaxial extrusion process to fabricate a core-shell hydrogel microfiber with high NSC density in the core portion. Oxidized hyaluronic acid, carboxymethyl chitosan, and matrigel blend were used as a matrix for NSC growth and to facilitate the fabrication process. During thein vitrodifferentiation culture, it was found that NSC microfibers could differentiate into neurons and astrocytes with higher efficiency compared to NSC cultured in petri dishes. Furthermore, duringin vivotransplantation, NSC microfibers were coated with polylactic acid nanosheets by electrospinning for reinforcement. The coated NSC nanofibers exhibited higher anti-inflammatory effect and lesion cavity filling rate compared with the control group. Meanwhile, more neuron- and oligodendrocyte-like cells were visualized at the lesion epicenter. Finally, axon regrowth across the whole lesion site was observed, demonstrating that the microfiber could guide renascent axon regrowth. Experiment results indicate that the NSC microfiber is a promising bioactive treatment for complete SCI treatment with superior outcomes.


Subject(s)
Axons , Cell Differentiation , Neural Stem Cells , Neurons , Spinal Cord Injuries , Tissue Scaffolds , Animals , Neural Stem Cells/drug effects , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Spinal Cord Injuries/therapy , Spinal Cord Injuries/pathology , Axons/drug effects , Axons/physiology , Axons/metabolism , Cell Differentiation/drug effects , Neurons/cytology , Neurons/drug effects , Tissue Scaffolds/chemistry , Rats, Sprague-Dawley , Hydrogels/chemistry , Hydrogels/pharmacology , Chitosan/chemistry , Chitosan/pharmacology , Chitosan/analogs & derivatives , Cells, Cultured , Nerve Regeneration/drug effects , Nanofibers/chemistry , Rats , Female
17.
J Neurosci Methods ; 407: 110144, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38670535

ABSTRACT

BACKGROUND: The enteric nervous system (ENS) is comprised of neurons, glia, and neural progenitor cells that regulate essential gastrointestinal functions. Advances in high-efficiency enteric neuron culture would facilitate discoveries surrounding ENS regulatory processes, pathophysiology, and therapeutics. NEW METHOD: Development of a simple, robust, one-step method to culture murine enteric neurospheres in a 3D matrix that supports neural growth and differentiation. RESULTS: Myenteric plexus cells isolated from the entire length of adult murine small intestine formed ≥3000 neurospheres within 7 days. Matrigel-embedded neurospheres exhibited abundant neural stem and progenitor cells expressing Sox2, Sox10 and Msi1 by day 4. By day 5, neural progenitor cell marker Nestin appeared in the periphery of neurospheres prior to differentiation. Neurospheres produced extensive neurons and neurites, confirmed by Tubulin beta III, PGP9.5, HuD/C, and NeuN immunofluorescence, including neural subtypes Calretinin, ChAT, and nNOS following 8 days of differentiation. Individual neurons within and external to neurospheres generated depolarization induced action potentials which were inhibited in the presence of sodium channel blocker, Tetrodotoxin. Differentiated neurospheres also contained a limited number of glia and endothelial cells. COMPARISON WITH EXISTING METHODS: This novel one-step neurosphere growth and differentiation culture system, in 3D format (in the presence of GDNF, EGF, and FGF2), allows for ∼2-fold increase in neurosphere count in the derivation of enteric neurons with measurable action potentials. CONCLUSION: Our method describes a novel, robust 3D culture of electrophysiologically active enteric neurons from adult myenteric neural stem and progenitor cells.


Subject(s)
Myenteric Plexus , Neurons , Animals , Myenteric Plexus/cytology , Myenteric Plexus/physiology , Neurons/physiology , Neurons/cytology , Neurons/drug effects , Cell Culture Techniques/methods , Neural Stem Cells/cytology , Neural Stem Cells/physiology , Neural Stem Cells/drug effects , Cell Differentiation/physiology , Cell Differentiation/drug effects , Mice , Mice, Inbred C57BL , Cells, Cultured , Action Potentials/physiology , Action Potentials/drug effects , Laminin/pharmacology , Drug Combinations , Proteoglycans/pharmacology , Male , Neurogenesis/physiology , Neurogenesis/drug effects , Collagen
18.
Phytomedicine ; 128: 155362, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38522312

ABSTRACT

BACKGROUND: Stroke is a leading cause of disability and death worldwide. Currently, there is a lack of clinically effective treatments for the brain damage following ischemic stroke. Catalpol is a bioactive compound derived from the traditional Chinese medicine Rehmannia glutinosa and shown to be protective in various neurological diseases. However, the potential roles of catalpol against ischemic stroke are still not completely clear. PURPOSE: This study aimed to further elucidate the protective effects of catalpol against ischemic stroke. METHODS: A rat permanent middle cerebral artery occlusion (pMCAO) and oxygen-glucose deprivation (OGD) model was established to assess the effect of catalpol in vivo and in vitro, respectively. Behavioral tests were used to examine the effects of catalpol on neurological function of ischemic rats. Immunostaining was performed to evaluate the proliferation, migration and differentiation of neural stem cells (NSCs) as well as the angiogenesis in each group. The protein level of related molecules was detected by western-blot. The effects of catalpol on cultured NSCs as well as brain microvascular endothelial cells (BMECs) subjected to OGD in vitro were also examined by similar methods. RESULTS: Catalpol attenuated the neurological deficits and improved neurological function of ischemic rats. It stimulated the proliferation of NSCs in the subventricular zone (SVZ), promoted their migration to the ischemic cortex and differentiation into neurons or glial cells. At the same time, catalpol increased the cerebral vessels density and the number of proliferating cerebrovascular endothelial cells in the infracted cortex of ischemic rats. The level of SDF-1α and CXCR4 in the ischemic cortex was found to be enhanced by catalpol treatment. Catalpol was also shown to promote the proliferation and migration of cultured NSCs as well as the proliferation of BMECs subjected to OGD insult in vitro. Interestingly, the impact of catalpol on cultured cells was inhibited by CXCR4 inhibitor AMD3100. Moreover, the culture medium of BMECs containing catalpol promoted the proliferation of NSCs, which was also suppressed by AMD3100. CONCLUSION: Our data demonstrate that catalpol exerts neuroprotective effects by promoting neurogenesis and angiogenesis via the SDF-1α/CXCR4 pathway, suggesting the therapeutic potential of catalpol in treating cerebral ischemia.


Subject(s)
Chemokine CXCL12 , Iridoid Glucosides , Ischemic Stroke , Neurogenesis , Rats, Sprague-Dawley , Receptors, CXCR4 , Rehmannia , Animals , Iridoid Glucosides/pharmacology , Receptors, CXCR4/metabolism , Neurogenesis/drug effects , Chemokine CXCL12/metabolism , Male , Rehmannia/chemistry , Ischemic Stroke/drug therapy , Infarction, Middle Cerebral Artery/drug therapy , Neural Stem Cells/drug effects , Cell Proliferation/drug effects , Rats , Neuroprotective Agents/pharmacology , Neovascularization, Physiologic/drug effects , Cell Movement/drug effects , Cell Differentiation/drug effects , Endothelial Cells/drug effects , Disease Models, Animal , Signal Transduction/drug effects , Cells, Cultured , Angiogenesis
19.
Neuron ; 112(9): 1426-1443.e11, 2024 May 01.
Article in English | MEDLINE | ID: mdl-38442714

ABSTRACT

Glucocorticoids are important for proper organ maturation, and their levels are tightly regulated during development. Here, we use human cerebral organoids and mice to study the cell-type-specific effects of glucocorticoids on neurogenesis. We show that glucocorticoids increase a specific type of basal progenitors (co-expressing PAX6 and EOMES) that has been shown to contribute to cortical expansion in gyrified species. This effect is mediated via the transcription factor ZBTB16 and leads to increased production of neurons. A phenome-wide Mendelian randomization analysis of an enhancer variant that moderates glucocorticoid-induced ZBTB16 levels reveals causal relationships with higher educational attainment and altered brain structure. The relationship with postnatal cognition is also supported by data from a prospective pregnancy cohort study. This work provides a cellular and molecular pathway for the effects of glucocorticoids on human neurogenesis that relates to lasting postnatal phenotypes.


Subject(s)
Cerebral Cortex , Glucocorticoids , Neurogenesis , Promyelocytic Leukemia Zinc Finger Protein , Neurogenesis/drug effects , Neurogenesis/physiology , Humans , Animals , Mice , Glucocorticoids/pharmacology , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Cerebral Cortex/cytology , Female , Promyelocytic Leukemia Zinc Finger Protein/metabolism , Pregnancy , Neurons/metabolism , Neurons/drug effects , Organoids/drug effects , Organoids/metabolism , Gene Expression Regulation, Developmental/drug effects , Neural Stem Cells/drug effects , Neural Stem Cells/metabolism , Male
20.
Adv Sci (Weinh) ; 11(21): e2308993, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38516757

ABSTRACT

Neural stem cells (NSCs) transplantation is an attractive and promising treatment strategy for spinal cord injury (SCI). Various pathological processes including the severe inflammatory cascade and difficulty in stable proliferation and differentiation of NSCs limit its application and translation. Here, a novel physico-chemical bifunctional neural stem cells delivery system containing magnetic nanoparticles (MNPs and methylprednisolone (MP) is designed to repair SCI, the former regulates NSCs differentiation through magnetic mechanical stimulation in the chronic phase, while the latter alleviates inflammatory response in the acute phase. The delivery system releases MP to promote microglial M2 polarization, inhibit M1 polarization, and reduce neuronal apoptosis. Meanwhile, NSCs tend to differentiate into functional neurons with magnetic mechanical stimulation generated by MNPs in the static magnetic field, which is related to the activation of the PI3K/AKT/mTOR pathway. SCI mice achieve better functional recovery after receiving NSCs transplantation via physico-chemical bifunctional delivery system, which has milder inflammation, higher number of M2 microglia, more functional neurons, and axonal regeneration. Together, this bifunctional NSCs delivery system combined physical mechanical stimulation and chemical drug therapy is demonstrated to be effective, which provides new treatment insights into clinical transformation of SCI repair.


Subject(s)
Disease Models, Animal , Magnetite Nanoparticles , Methylprednisolone , Neural Stem Cells , Spinal Cord Injuries , Animals , Spinal Cord Injuries/therapy , Methylprednisolone/pharmacology , Mice , Neural Stem Cells/transplantation , Neural Stem Cells/drug effects , Magnetite Nanoparticles/chemistry , Magnetite Nanoparticles/therapeutic use , Cell Differentiation/drug effects , Stem Cell Transplantation/methods
SELECTION OF CITATIONS
SEARCH DETAIL
...