Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 39
1.
Environ Int ; 186: 108643, 2024 Apr.
Article En | MEDLINE | ID: mdl-38615544

Exposure to bisphenol S (BPS) is known to adversely affect neuronal development. As pivotal components of neuronal polarization, axons and dendrites are indispensable structures within neurons, crucial for the maintenance of nervous system function. Here, we investigated the impact of BPS exposure on axonal and dendritic development both in vivo and in vitro. Our results revealed that exposure to BPS during pregnancy and lactation led to a reduction in the complexity, density, and length of axons and dendrites in the prefrontal cortex (PFC) of offspring. Employing RNA sequencing technology to elucidate the underlying mechanisms of axonal and dendritic damage induced by BPS, Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis highlighted a significant alteration in the oxidative phosphorylation (OXPHOS) pathway, essential for mitochondrial function. Subsequent experiments demonstrate BPS-induced impairment in mitochondrial function, including damaged morphology, decreased adenosine triphosphate (ATP) and superoxide dismutase (SOD) levels, and increased reactive oxygen species and malondialdehyde (MDA). These alterations coincided with the downregulated expression of OXPHOS pathway-related genes (ATP6V1B1, ATP5K, NDUFC1, NDUFC2, NDUFA3, COX6B1) and Myosin 19 (Myo19). Notably, Myo19 overexpression restored the BPS-induced mitochondrial dysfunction by alleviating the inhibition of OXPHOS pathway. Consequently, this amelioration was associated with a reduction in BPS-induced axonal and dendritic injury observed in cultured neurons of the PFC.


Axons , Dendrites , Mitochondria , Oxidative Phosphorylation , Phenols , Sulfones , Animals , Mitochondria/drug effects , Mitochondria/metabolism , Phenols/toxicity , Dendrites/drug effects , Oxidative Phosphorylation/drug effects , Female , Sulfones/toxicity , Axons/drug effects , Pregnancy , Prefrontal Cortex/drug effects , Prefrontal Cortex/metabolism , Mice
2.
Environ Pollut ; 347: 123740, 2024 Apr 15.
Article En | MEDLINE | ID: mdl-38462198

Tris (1,3-dichloro-2-propyl) phosphate (TDCPP), a halogen-containing phosphorus flame retardant, is widely used and has been shown to possess health risks to humans. The sustained release of artificial nanomaterials into the environment increases the toxicological risks of their coexisting pollutants. Nanomaterials may seriously change the environmental behavior and fate of pollutants. In this study, we investigated this combined toxicity and the potential mechanisms of toxicity of TDCPP and titanium dioxide nanoparticles (TiO2 NPs) aggregates on human neuroblastoma SH-SY5Y cells. TDCPP and TiO2 NPs aggregates were exposed in various concentration combinations, revealing that TDCPP (25 µg/mL) reduced cell viability, while synergistic exposure to TiO2 NPs aggregates exacerbated cytotoxicity. This combined exposure also disrupted mitochondrial function, leading to dysregulation in the expression of mitochondrial fission proteins (DRP1 and FIS1) and fusion proteins (OPA1 and MFN1). Consequently, excessive mitochondrial fission occurred, facilitating the translocation of cytochrome C from mitochondria to activate apoptotic signaling pathways. Furthermore, exposure of the combination of TDCPP and TiO2 NPs aggregates activated upstream mitochondrial autophagy but disrupted downstream Parkin recruitment to damaged mitochondria, preventing autophagosome-lysosome fusion and thereby disrupting mitochondrial autophagy. Altogether, our findings suggest that TDCPP and TiO2 NPs aggregates may stimulate apoptosis in neuronal SH-SY5Y cells by inducing mitochondrial hyperfission and inhibiting mitochondrial autophagy.


Environmental Pollutants , Neuroblastoma , Humans , Mitophagy , Neuroblastoma/metabolism , Mitochondrial Dynamics , Apoptosis
3.
Ecotoxicol Environ Saf ; 271: 115991, 2024 Feb.
Article En | MEDLINE | ID: mdl-38237395

Heavy metal toxicity is a significant global health concern, with particular attention given to lead (Pb) exposure due to its adverse effects on cognitive development, especially in children exposed to low concentrations. While Pb neurotoxicity has been extensively studied, the analysis and molecular mechanisms underlying the transgenerational effects of Pb exposure-induced neurotoxicity remain poorly understood. In this study, we utilized Drosophila, a powerful developmental animal model, to investigate this phenomenon. Our findings demonstrated that Pb exposure during the developmental stage had a profound effect on the neurodevelopment of F0 fruit flies. Specifically, we observed a loss of correlation between the terminal motor area and muscle fiber area, along with an increased frequency of the ß-lobe midline crossing phenotype in mushroom bodies. Western blot analysis indicated altered expression levels of synaptic vesicle proteins, with a decrease in Synapsin (SYN) and an increase in Bruchpilot (BRP) expression, suggesting changes in synaptic vesicle release sites. These findings were corroborated by electrophysiological data, showing an increase in the amplitude of evoked excitatory junctional potential (EJP) and an increase in the frequency of spontaneous excitatory junctional potential (mEJP) following Pb exposure. Importantly, our results further confirmed that the developmental neurotoxicity resulting from grandparental Pb exposure exhibited a transgenerational effect. The F3 offspring displayed neurodevelopmental defects, synaptic function abnormalities, and repetitive behavior despite lacking direct Pb exposure. Our MeDIP-seq analysis further revealed significant alterations in DNA methylation levels in several neurodevelopmental associated genes (eagle, happyhour, neuroglian, bazooka, and spinophilin) in the F3 offspring exposed to Pb. These findings suggest that DNA methylation modifications may underlie the inheritance of acquired phenotypic traits resulting from environmental Pb exposure.


Drosophila melanogaster , Neurotoxicity Syndromes , Animals , Child , Humans , Lead/metabolism , DNA Methylation , Neurotoxicity Syndromes/genetics , Genome
4.
Int J Nanomedicine ; 18: 7183-7204, 2023.
Article En | MEDLINE | ID: mdl-38076727

The increasing use of titanium dioxide nanoparticles (TiO2 NPs) across various fields has led to a growing concern regarding their environmental contamination and inevitable human exposure. Consequently, significant research efforts have been directed toward understanding the effects of TiO2 NPs on both humans and the environment. Notably, TiO2 NPs exposure has been associated with multiple impairments of the nervous system. This review aims to provide an overview of the documented neurotoxic effects of TiO2 NPs in different species and in vitro models. Following exposure, TiO2 NPs can reach the brain, although the specific mechanism and quantity of particles that cross the blood-brain barrier (BBB) remain unclear. Exposure to TiO2 NPs has been shown to induce oxidative stress, promote neuroinflammation, disrupt brain biochemistry, and ultimately impair neuronal function and structure. Subsequent neuronal damage may contribute to various behavioral disorders and play a significant role in the onset and progression of neurodevelopmental or neurodegenerative diseases. Moreover, the neurotoxic potential of TiO2 NPs can be influenced by various factors, including exposure characteristics and the physicochemical properties of the TiO2 NPs. However, a systematic comparison of the neurotoxic effects of TiO2 NPs with different characteristics under various exposure conditions is still lacking. Additionally, our understanding of the underlying neurotoxic mechanisms exerted by TiO2 NPs remains incomplete and fragmented. Given these knowledge gaps, it is imperative to further investigate the neurotoxic hazards and risks associated with exposure to TiO2 NPs.


Metal Nanoparticles , Nanoparticles , Neurotoxicity Syndromes , Humans , Nanoparticles/toxicity , Nanoparticles/chemistry , Oxidative Stress , Titanium/chemistry , Brain , Neurotoxicity Syndromes/etiology , Metal Nanoparticles/toxicity , Metal Nanoparticles/chemistry
5.
Commun Biol ; 6(1): 1056, 2023 10 18.
Article En | MEDLINE | ID: mdl-37853189

Neuroligins are transmembrane cell adhesion proteins well-known for their genetic links to autism spectrum disorders. Neuroligins can function by regulating the actin cytoskeleton, however the factors and mechanisms involved are still largely unknown. Here, using the Drosophila neuromuscular junction as a model, we reveal that F-Actin assembly at the Drosophila NMJ is controlled through Cofilin signaling mediated by an interaction between DNlg2 and RACK1, factors not previously known to work together. The deletion of DNlg2 displays disrupted RACK1-Cofilin signaling pathway with diminished actin cytoskeleton proteo-stasis at the terminal of the NMJ, aberrant NMJ structure, reduced synaptic transmission, and abnormal locomotion at the third-instar larval stage. Overexpression of wildtype and activated Cofilin in muscles are sufficient to rescue the morphological and physiological defects in dnlg2 mutants, while inactivated Cofilin is not. Since the DNlg2 paralog DNlg1 is known to regulate F-actin assembly mainly via a specific interaction with WAVE complex, our present work suggests that the orchestration of F-actin by Neuroligins is a diverse and complex process critical for neural connectivity.


Drosophila Proteins , Drosophila , Animals , Drosophila/genetics , Drosophila/metabolism , Actin Depolymerizing Factors/genetics , Actin Depolymerizing Factors/metabolism , Actins/metabolism , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Signal Transduction/physiology , Receptors for Activated C Kinase/genetics
7.
Ecotoxicol Environ Saf ; 260: 115098, 2023 Jul 15.
Article En | MEDLINE | ID: mdl-37269611

As a result of the ban on bisphenol A (BPA), a hormone disruptor with developmental neurotoxicity, several BPA derivatives (BPs) have been widely used in industrial production. However, there are no effective methods for assessing the neurodevelopmental toxic effects of BPs. To address this, a Drosophila exposure model was established, and W1118 was reared in food containing these BPs. Results showed that each BPs displayed different semi-lethal doses ranging from 1.76 to 19.43 mM. Exposure to BPs delayed larval development and affected axonal growth, resulting in the abnormal crossing of the midline of axons in the ß lobules of mushroom bodies, but the damage caused by BPE and BPF was relatively minor. BPC, BPAF, and BPAP have the most significant effects on locomotor behavior, whereas BPC exhibited the most affected social interactions. Furthermore, exposure to high-dose BPA, BPC, BPS, BPAF, and BPAP also significantly increased the expression of Drosophila estrogen-related receptors. These demonstrated that different kinds of BPs had different levels of neurodevelopmental toxicity, and the severity was BPZ > BPC and BPAF > BPB > BPS > BPAP ≈ BPAl ≈ BPF > BPE. Therefore, BPZ, BPC, BPS, BPAF, and BPAP should be evaluated as potential alternatives to BPA.


Benzhydryl Compounds , Drosophila melanogaster , Animals , Benzhydryl Compounds/toxicity , Phenols/toxicity , Food
8.
Sci Total Environ ; 888: 164076, 2023 Aug 25.
Article En | MEDLINE | ID: mdl-37196938

Titanium dioxide nanoparticles (TiO2 NPs) are widely used in several consumer products. However, because of their neurotoxic nature, exposure to TiO2 NPs could impair locomotor behavior. Whether the impairment in locomotor behavior caused by TiO2 NPs exposure is sustained and the effects is gender-specific has remained elusive, warranting further studies to elucidate the underlying mechanisms. Thus, we established a Drosophila model to study the effects of chronic TiO2 NPs exposure on the locomotor behavior of Drosophila in different generations and explore the underlying mechanisms. Chronic TiO2 NPs exposure caused accumulation of Ti in the body and affected the life history traits of Drosophila. Furthermore, chronic exposure to TiO2 NPs decreased the total crawling distance of larvae and the total movement distance of adult males in the F3 generation, indicating the damage caused to the locomotor behavior of Drosophila. Impaired neuromuscular junction (NMJ) morphology was observed, manifested by the reduced number of boutons, size of boutons, and branch length of NMJ. In addition, several differentially expressed genes (DEGs) related to NMJ development were selected by RNA sequencing and their expression was confirmed by quantitative real-time-polymerase chain reaction (qRT-PCR). Compared with the control group, the gene expression of Cyp6a17, frac, and kek2 in the TiO2 NPs exposure group decreased, whereas that of Gba1a, Hll and List was elevated. These findings indicated that chronic TiO2 NPs exposure damage the morphology of NMJ by altering the expression of genes related to NMJ development, consequently causing locomotor behavior deficits in Drosophila.


Metal Nanoparticles , Nanoparticles , Male , Animals , Drosophila , Nanoparticles/toxicity , Titanium/toxicity , Larva/metabolism , Metal Nanoparticles/toxicity
9.
Int J Mol Sci ; 24(3)2023 Jan 17.
Article En | MEDLINE | ID: mdl-36768153

Autism spectrum disorder (ASD) is a heterogeneous, behaviorally defined neurodevelopmental disorder. Over the past two decades, the prevalence of autism spectrum disorders has progressively increased, however, no clear diagnostic markers and specifically targeted medications for autism have emerged. As a result, neurobehavioral abnormalities, neurobiological alterations in ASD, and the development of novel ASD pharmacological therapy necessitate multidisciplinary collaboration. In this review, we discuss the development of multiple animal models of ASD to contribute to the disease mechanisms of ASD, as well as new studies from multiple disciplines to assess the behavioral pathology of ASD. In addition, we summarize and highlight the mechanistic advances regarding gene transcription, RNA and non-coding RNA translation, abnormal synaptic signaling pathways, epigenetic post-translational modifications, brain-gut axis, immune inflammation and neural loop abnormalities in autism to provide a theoretical basis for the next step of precision therapy. Furthermore, we review existing autism therapy tactics and limits and present challenges and opportunities for translating multidisciplinary knowledge of ASD into clinical practice.


Autism Spectrum Disorder , Autistic Disorder , Animals , Autism Spectrum Disorder/genetics , Autism Spectrum Disorder/therapy , Autistic Disorder/genetics , Risk Factors , Inflammation , Biological Factors/therapeutic use
10.
Brain Behav Immun ; 103: 85-96, 2022 07.
Article En | MEDLINE | ID: mdl-35427759

Recent progress on the central lymphatic system has greatly increased our understanding of how the brain maintains its own waste homeostasis. Here, we showed that perivascular spaces and meningeal lymphatic vessels form a functional route for clearance of senescent astrocytes from the aging brain. Blocking meningeal lymphatic drainage by ligation of the deep cervical lymph nodes impaired clearance of senescent astrocytes from brain parenchyma, subsequently increasing neuroinflammation in aged mice. By contrast, enhancing meningeal lymphatic vessel diameter by a recombinant adeno-associated virus encoding mouse vascular endothelial growth factor-C (VEGF-C) improved clearance of senescent astrocytes and mitigated neuroinflammation. Mechanistically, VEGF-C was highly expressed in senescent astrocytes, contributing themselves to migrate across lymphatic vessels along C-C motif chemokine ligand 21 (CCL21) gradient by interacting with VEGF receptor 3. Moreover, intra-cisternal injection of antibody against CCL21 hampered senescent astrocytes into the lymphatic vessels and exacerbated short memory defects of aged mice. Together, these findings reveal a new perspective for the meningeal lymphatics in the removal of senescent astrocytes, thus offering a valuable target for therapeutic intervention.


Lymphatic Vessels , Vascular Endothelial Growth Factor C , Animals , Astrocytes/metabolism , Brain/metabolism , Lymphatic System , Lymphatic Vessels/metabolism , Mice , Vascular Endothelial Growth Factor C/metabolism
11.
Biochem Biophys Res Commun ; 525(2): 469-476, 2020 04 30.
Article En | MEDLINE | ID: mdl-32107003

AIM: The fragile X mental retardation protein (FMRP), the product of the FMR1 gene, is responsible for the fragile X syndrome (FXS). FMRP regulates miRNA expression and is involved in miRNA-mediated gene silencing. However, the question of whether FMRP is, in turn, regulated by miRNAs remains unanswered. MAIN METHODS: We detected the FMRP expression pattern by in situ hybridization. MiR-315 overexpression and knockout models were generated by germ-line transformation and ends-out homologous recombination, respectively. Western blotting and immunohistochemistry were used to detect Drosophila FMRP (dFMRP) and a Luciferase reporter assay was used to confirm the regulation of dfmr1 mRNA by mir-315. Synaptic structural quantification and electrophysiological methods were used to compare synaptic functions among groups. KEY FINDINGS: Here, we determined that the transcription product of dFMR1, the Drosophila homologue of FMR1, is a direct target of miR-315. MiR-315 is mainly expressed in the nervous system of Drosophila. Flies overexpressing miR-315 showed pupation defects and reduced hatching rates. A homozygous miR-315 knockout status is embryonic lethal in flies. These observations indicate that miR-315 is a key regulator of the Drosophila nervous system. Furthermore, computational prediction and cell-based luciferase and in vivo assays demonstrated that dfmr1 is directly targeted by miR-315. Lastly, using the neuromuscular junction as a model, we found that miR-315 regulates synaptic structure and transmission by targeting dfmr1. SIGNIFICANCE: These findings provide compelling evidence that miR-315 targets dfmr1 in the Drosophila nervous system, acting as a regulatory factor for the fine-tuned modulation of FMRP expression.


Drosophila Proteins/genetics , Drosophila melanogaster/embryology , Drosophila melanogaster/genetics , Fragile X Mental Retardation Protein/genetics , Gene Expression Regulation, Developmental , Animals , Nervous System/embryology , Nervous System/metabolism , Neurogenesis , Neuromuscular Junction/genetics , Synapses/genetics
12.
Ultrason Sonochem ; 62: 104875, 2020 Apr.
Article En | MEDLINE | ID: mdl-31796329

Episodic release of bioactive compounds is often necessary for appropriate biological effects under specific physiological conditions. Here, we aimed to develop an injectable, biocompatible, and thermosensitive hydrogel system for ultrasound (US)-triggered drug release. An mPEG-PLGA-BOX block copolymer hydrogel was synthesized. The viscosity of 15 wt% hydrogel is 0.03 Pa*s at 25 °C (liquid form) and 34.37 Pa*s at 37 °C (gel form). Baseline and US-responsive in vitro release profile of a small molecule (doxorubicin) and that of a large molecule (FITC-dextran), from the hydrogel, was tested. A constant baseline release was observed in vitro for 7 d. When triggered by US (1 MHz, continuous, 0.4 W/cm2), the release rate increased by approximately 70 times. Without US, the release rate returned to baseline. Baseline and US-responsive in vivo release profile of doxorubicin was tested by subcutaneous injection in the back of mice and rats. Following injection into the subcutaneous layer, in vivo results also suggested that the hydrogels remained in situ and provided a steady release for at least 7 d; in the presence of the US-trigger, in vivo release from the hydrogel increased by approximately 10 times. Therefore, the mPEG-PLGA-BOX block copolymer hydrogel may serve as an injectable, biocompatible, and thermosensitive hydrogel system that is applicable for US-triggered drug release.


Biocompatible Materials , Delayed-Action Preparations , Dextrans/administration & dosage , Doxorubicin/administration & dosage , Fluorescein-5-isothiocyanate/analogs & derivatives , Hydrogels , Ultrasonic Waves , Animals , Cell Line , Fluorescein-5-isothiocyanate/administration & dosage , Injections, Subcutaneous , Mice , Polyesters/chemistry , Polyethylene Glycols/chemistry , Rats
13.
Autophagy ; 16(4): 672-682, 2020 04.
Article En | MEDLINE | ID: mdl-31242080

A shared neuropathological hallmark in amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD) is nuclear clearance and cytoplasmic aggregation of TARDBP/TDP-43 (TAR DNA binding protein). We previously showed that the ability of TARDBP to repress nonconserved cryptic exons was impaired in brains of patients with ALS and FTD, suggesting that its nuclear depletion contributes to neurodegeneration. However, the critical pathways impacted by the failure to repress cryptic exons that may contribute to neurodegeneration remain undefined. Here, we report that transcriptome analysis of TARDBP-deficient neurons revealed downregulation of ATG7, a critical gene required for macroautophagy/autophagy. Mouse and Drosophila models lacking TARDBP/TBPH in motor neurons exhibiting age-dependent neurodegeneration and motor deficits showed reduction of ATG7 and accumulation of SQSTM1/p62 inclusions. Importantly, genetic upregulation of the autophagy pathway improved motor function and survival in TBPH-deficient flies. Together with our observation that ATG7 is reduced in ALS-FTD brain tissues, these findings identify the autophagy pathway as one key effector of nuclear depletion of TARDBP that contributes to neurodegeneration. We thus suggest that the autophagy pathway is a therapeutic target for ALS-FTD and other disorders exhibiting TARDBP pathology.Abbreviations: ALS: amyotrophic lateral sclerosis; ANOVA: analysis of variance; ChAT: choline acetyltransferase; CTSD: cathepsin D; FTD: frontotemporal dementia; LAMP1: lysosomal associated membrane protein 1; NMJ: neuromuscular junction; RBFOX3/NeuN: RNA binding fox-1 homolog 3; SQSTM1: sequestosome 1; TARDBP/TDP-43: TAR DNA binding protein 43.


Autophagy-Related Protein 7/metabolism , DNA-Binding Proteins/metabolism , Motor Neurons/metabolism , Animals , Autophagy/genetics , Autophagy/physiology , Brain/metabolism , Humans , Mice, Transgenic , Motor Neurons/pathology , Up-Regulation
14.
Acta Neuropathol ; 138(5): 813-826, 2019 11.
Article En | MEDLINE | ID: mdl-31332509

Nuclear depletion of TDP-43, an essential RNA binding protein, may underlie neurodegeneration in amyotrophic lateral sclerosis (ALS). As several functions have been ascribed to this protein, the critical role(s) of TDP-43 in motor neurons that may be compromised in ALS remains unknown. We show here that TDP-43 mediated splicing repression, which serves to protect the transcriptome by preventing aberrant splicing, is central to the physiology of motor neurons. Expression in Drosophila TDP-43 knockout models of a chimeric repressor, comprised of the RNA recognition domain of TDP-43 fused to an unrelated splicing repressor, RAVER1, attenuated motor deficits and extended lifespan. Likewise, AAV9-mediated delivery of this chimeric rescue repressor to mice lacking TDP-43 in motor neurons delayed the onset, slowed the progression of motor symptoms, and markedly extended their lifespan. In treated mice lacking TDP-43 in motor neurons, aberrant splicing was significantly decreased and accompanied by amelioration of axon degeneration and motor neuron loss. This AAV9 strategy allowed long-term expression of the chimeric repressor without any adverse effects. Our findings establish that splicing repression is a major function of TDP-43 in motor neurons and strongly support the idea that loss of TDP-43-mediated splicing fidelity represents a key pathogenic mechanism underlying motor neuron loss in ALS.


DNA-Binding Proteins/genetics , Motor Neurons/pathology , Nerve Degeneration/genetics , RNA Splicing/genetics , Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/pathology , Animals , Cell Nucleus/metabolism , DNA-Binding Proteins/metabolism , Drosophila , Humans , Motor Neurons/metabolism , Nerve Degeneration/pathology , RNA-Binding Proteins/metabolism
16.
Parkinsons Dis ; 2019: 9193534, 2019.
Article En | MEDLINE | ID: mdl-30944722

7,8-Dihydroxyflavone (7,8-DHF) is thought to be a promising therapeutic agent for various neurodegenerative diseases. The major purpose of this study was to investigate the neuroprotective effects of 7,8-DHF on the rotenone-induced motor deficit of Parkinson's disease. Nine-month-old rats were treated with rotenone (2 mg/kg/day, i.h.) for 5 weeks to establish the animal model of Parkinson's disease (PD), and 7,8-DHF (5 mg/kg, i.p.) was administrated daily throughout the whole period of rotenone injection. Five weeks later, an open field test was used to assess the motor ability of the animals. TH immunostaining was performed to evaluate rotenone-induced neurotoxicity on substantia nigra (SN) dopaminergic neurons and the DA terminals in the striatum. Western blot analyses were used to examine the expressions of TH, BDNF/TrkB signaling cascades, phospho-α-synuclein (Ser129), α-synuclein, and phospho-tau (Ser396) in SN. The results revealed that treatment with 7,8-DHF improved PD model's behavioral performance and reduced dopaminergic neuron loss in the SN and striatum, associated with the activation of TrkB receptors and its signaling cascades, and reduced p-MAPK, p-α-synuclein, and p-tau. Collectively, these results indicated that 7,8-DHF displayed prominent neuroprotective properties, providing a promising therapeutic strategy for PD treatment.

17.
Exp Cell Res ; 374(2): 342-352, 2019 01 15.
Article En | MEDLINE | ID: mdl-30553967

Guanine nucleotide exchange factors (GEFs) are essential for small G proteins to activate their downstream signaling pathways, which are involved in morphogenesis, cell adhesion, and migration. Mutants of Gef26, a PDZ-GEF (PDZ domain-containing guanine nucleotide exchange factor) in Drosophila, exhibit strong defects in wings, eyes, and the reproductive and nervous systems. However, the precise roles of Gef26 in development remain unclear. In the present study, we analyzed the role of Gef26 in synaptic development and function. We identified significant decreases in bouton number and branch length at larval neuromuscular junctions (NMJs) in Gef26 mutants, and these defects were fully rescued by restoring Gef26 expression, indicating that Gef26 plays an important role in NMJ morphogenesis. In addition to the observed defects in NMJ morphology, electrophysiological analyses revealed functional defects at NMJs, and locomotor deficiency appeared in Gef26 mutant larvae. Furthermore, Gef26 regulated NMJ morphogenesis by regulating the level of synaptic Fasciclin II (FasII), a well-studied cell adhesion molecule that functions in NMJ development and remodeling. Finally, our data demonstrate that Gef26-specific small G protein Rap1 worked downstream of Gef26 to regulate the level of FasII at NMJs, possibly through a ßPS integrin-mediated signaling pathway. Taken together, our findings define a novel role of Gef26 in regulating NMJ development and function.


Drosophila Proteins/metabolism , Drosophila/metabolism , Neuromuscular Junction/metabolism , Synapses/metabolism , Synaptic Transmission/physiology , Telomere-Binding Proteins/metabolism , Animals , Cell Adhesion/physiology , Cell Adhesion Molecules, Neuronal/metabolism , Guanine Nucleotide Exchange Factors/metabolism , Larva/metabolism , Presynaptic Terminals/metabolism , Shelterin Complex , Signal Transduction/physiology , Synapses/physiology
18.
Sci Rep ; 8(1): 2218, 2018 02 02.
Article En | MEDLINE | ID: mdl-29396523

Despite extensive efforts in recent years, the blood-brain barrier (BBB) remains a significant obstacle for drug delivery. This study proposes using a clinical extracorporeal shockwave instrument to open the BBB, combined with a laser assisted bi-axial locating platform to achieve non-invasive, controllable-focus and reversible BBB opening in the brains of rats. Under shockwave treatment with an intensity level of 5 (P-9.79 MPa, energy flux density (EFD) 0.21 mJ/mm2) and a pulse repetition frequency of 5 Hz, the BBB could be opened after 50 shocks without the use of an ultrasound contrast agent. With the proposed method, the BBB opening can be precisely controlled in terms of depth, size and location. Moreover, a shockwave based gene transfection was demonstrated using a luciferase gene.


Blood-Brain Barrier/radiation effects , Extracorporeal Shockwave Therapy/methods , Genetic Therapy/methods , High-Energy Shock Waves , Transfection , Animals , Brain/radiation effects , Genes, Reporter , Luciferases/analysis , Luciferases/genetics , Rats
19.
Ultrasonics ; 83: 157-163, 2018 Feb.
Article En | MEDLINE | ID: mdl-28408049

Episodic release of bioactive compounds plays an important role in biological systems. "On-demand" release systems which based on polymeric materials and activated by external stimuli may provide the necessary functionality. Here we describe an ultrasound-responsive hydrogel based on N-isopropylacrylamide (NIPAM) and N,N'-methylenebisacrylamide (MBAm), which is suitable for triggered release of two large molecules: bovine serum albumin (BSA, 66kDa) and dextran (3-5kDa). It is shown that the release amount of these two large molecules increased with increasing hydrogel temperature, and the application of ultrasound further increased the release. By simply adjusting the contents of NIPAM and MBAm, the difference of BSA release between the presence and absence of ultrasound could be adjusted from 2.7 to 84 folds. There was also a positive correlation between the ultrasound intensity and release amount. These properties made the NIPAM-based hydrogel a tunable platform for focal drug delivery.

20.
J Vis Exp ; (125)2017 07 04.
Article En | MEDLINE | ID: mdl-28715400

Chronic Traumatic Encephalopathy (CTE) is an established neurodegenerative disease that is closely associated with exposure to repetitive mild Traumatic Brain Injury (mTBI). The mechanisms responsible for its complex pathological changes remain largely elusive, despite a recent consensus to define the neuropathological criteria. Here, we describe a novel method to develop a model of CTE in Drosophila melanogaster (Drosophila ) in an attempt to identify the key genes and pathways that lead to the characteristic hyperphosphorylated tau accumulation and neuronal death in the brain. Adjustable-strength impacts to inflict mild closed injury are delivered directly to the fly head, subjecting the head to rapid acceleration and deceleration. Our method eliminates the potential problems inherent with other Drosophila mTBI models (e.g.,animal death might be induced by damage to other parts of the body or to internal organs). The less labor- and cost-intensive animal care, short life span, and extensive genetic tools make the fruit fly ideal to study CTE pathogenesis and make it possible to perform large-scale, genome-wide forward genetic and pharmacological screens. We anticipate that the ongoing characterization of the model will generate important mechanistic insights on disease prevention and therapeutic approaches.


Brain Injuries/diagnosis , Brain/pathology , Chronic Traumatic Encephalopathy/diagnosis , Drosophila/growth & development , Neurodegenerative Diseases/diagnosis , Animals
...