Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 39
Filter
Add more filters










Publication year range
1.
PLoS One ; 17(5): e0265866, 2022.
Article in English | MEDLINE | ID: mdl-35588133

ABSTRACT

A major concern in tissue biopsies with a needle is missing the most lethal clone of a tumor, leading to a false negative result. This concern is well justified, since needle-based biopsies gather tissue information limited to needle size. In this work, we show that molecular harvesting with electroporation, e-biopsy, could increase the sampled tissue volume in comparison to tissue sampling by a needle alone. Suggested by numerical models of electric fields distribution, the increased sampled volume is achieved by electroporation-driven permeabilization of cellular membranes in the tissue around the sampling needle. We show that proteomic profiles, sampled by e-biopsy from the brain tissue, ex vivo, at 0.5mm distance outside the visible margins of mice brain melanoma metastasis, have protein patterns similar to melanoma tumor center and different from the healthy brain tissue. In addition, we show that e-biopsy probed proteome signature differentiates between melanoma tumor center and healthy brain in mice. This study suggests that e-biopsy could provide a novel tool for a minimally invasive sampling of molecules in tissue in larger volumes than achieved with traditional needle biopsies.


Subject(s)
Melanoma , Proteome , Animals , Brain/pathology , Electroporation , Margins of Excision , Melanoma/pathology , Mice , Proteomics
2.
Biomedicines ; 10(2)2022 Jan 29.
Article in English | MEDLINE | ID: mdl-35203533

ABSTRACT

BACKGROUND: Spinal cord injury (SCI) pathology includes both primary and secondary events. The primary injury includes the original traumatic event, and the secondary injury, beginning immediately after the initial injury, involves progressive neuroinflammation, neuronal excitotoxicity, gliosis, and degeneration. Currently, there is no effective neuroprotective treatment for SCI. However, an accumulating body of data suggests that PELF-EMF has beneficial therapeutic effects on neurotrauma. The purpose of this study was to test the efficacy of the PELF-EMF SEQEX device using a compression SCI mouse model. METHODS: C57BL/6 mice were exposed to PELF-EMF for 4 h on a daily basis for two months, beginning 2 h after a mild-moderate compression SCI. RESULTS: The PELF-EMF treatment significantly diminished inflammatory cell infiltration and astrocyte activation by reducing Iba1, F4/80, CD68+ cells, and GAFP at the lesion borders, and increased pro-survival signaling, such as BDNF, on the neuronal cells. Moreover, the treatment exhibited a neuroprotective effect by reducing the demyelination of the axons of the white matter at the lesion's center. CONCLUSIONS: Treatment with SEQEX demonstrated significant anti-inflammatory and neuroprotective effects. Considering our results, this safe and effective rehabilitative device, already available on the market, may provide a major therapeutic asset in the treatment of SCI.

3.
Adv Sci (Weinh) ; 9(11): e2105694, 2022 04.
Article in English | MEDLINE | ID: mdl-35128819

ABSTRACT

Cell therapy using induced pluripotent stem cell-derived neurons is considered a promising approach to regenerate the injured spinal cord (SC). However, the scar formed at the chronic phase is not a permissive microenvironment for cell or biomaterial engraftment or for tissue assembly. Engineering of a functional human neuronal network is now reported by mimicking the embryonic development of the SC in a 3D dynamic biomaterial-based microenvironment. Throughout the in vitro cultivation stage, the system's components have a synergistic effect, providing appropriate cues for SC neurogenesis. While the initial biomaterial supported efficient cell differentiation in 3D, the cells remodeled it to provide an inductive microenvironment for the assembly of functional SC implants. The engineered tissues are characterized for morphology and function, and their therapeutic potential is investigated, revealing improved structural and functional outcomes after acute and chronic SC injuries. Such technology is envisioned to be translated to the clinic to rewire human injured SC.


Subject(s)
Induced Pluripotent Stem Cells , Spinal Cord Injuries , Biocompatible Materials/chemistry , Humans , Neurons , Spinal Cord Injuries/therapy
4.
Sci Rep ; 11(1): 14644, 2021 07 19.
Article in English | MEDLINE | ID: mdl-34282238

ABSTRACT

Inhibition of extracellular glutamate (Glu) release decreases proliferation and invasion, induces apoptosis, and inhibits melanoma metastatic abilities. Previous studies have shown that Blood-glutamate scavenging (BGS), a novel treatment approach, has been found to be beneficial in attenuating glioblastoma progression by reducing brain Glu levels. Therefore, in this study we evaluated the ability of BGS treatment to inhibit brain metastatic melanoma progression in-vivo. RET melanoma cells were implanted in C56BL/6J mice to induce brain melanoma tumors followed by treatment with BGS or vehicle administered for fourteen days. Bioluminescent imaging was conducted to evaluate tumor growth, and plasma/CSF Glu levels were monitored throughout. Immunofluorescence staining of Ki67 and 53BP1 was used to analyze tumor cell proliferation and DNA double-strand breaks. In addition, we analyzed CD8, CD68, CD206, p-STAT1 and iNOS expression to evaluate alterations in tumor micro-environment and anti-tumor immune response due to treatment. Our results show that BGS treatment reduces CSF Glu concentration and consequently melanoma growth in-vivo by decreasing tumor cell proliferation and increasing pro-apoptotic signaling in C56BL/6J mice. Furthermore, BGS treatment supported CD8+ cell recruitment and CD68+ macrophage invasion. These findings suggest that BGS can be of potential therapeutic relevance in the treatment of metastatic melanoma.


Subject(s)
Aspartate Aminotransferase, Cytoplasmic/administration & dosage , Brain Neoplasms/drug therapy , Glutamic Acid/metabolism , Melanoma/drug therapy , Oxaloacetic Acid/administration & dosage , Animals , Apoptosis/drug effects , Aspartate Aminotransferase, Cytoplasmic/pharmacology , Brain Neoplasms/pathology , Brain Neoplasms/secondary , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Therapy, Combination , Glioblastoma/drug therapy , Glioblastoma/pathology , Glioblastoma/secondary , Humans , Melanoma/pathology , Mice , Mice, Inbred C57BL , Molecular Targeted Therapy/methods , Oxaloacetic Acid/pharmacology , Recombinant Proteins/administration & dosage , Signal Transduction/drug effects , Tumor Microenvironment/drug effects
5.
Transl Psychiatry ; 10(1): 341, 2020 Oct 06.
Article in English | MEDLINE | ID: mdl-33024083

ABSTRACT

An amendment to this paper has been published and can be accessed via a link at the top of the paper.

6.
Transl Psychiatry ; 10(1): 305, 2020 09 01.
Article in English | MEDLINE | ID: mdl-32873780

ABSTRACT

Schizophrenia is a debilitating psychiatric disorder with a significant number of patients not adequately responding to treatment. Phencyclidine (PCP) is used as a validated model for schizophrenia, shown to reliably induce positive, negative and cognitive-like behaviors in rodents. It was previously shown in our lab that behavioral phenotypes of PCP-treated mice can be alleviated after intracranial transplantation of mesenchymal stem cells (MSC). Here, we assessed the feasibility of intranasal delivery of MSCs-derived-extracellular vesicles (EVs) to alleviate schizophrenia-like behaviors in a PCP model of schizophrenia. As MSCs-derived EVs were already shown to concentrate at the site of lesion in the brain, we determined that in PCP induced injury the EVs migrate to the prefrontal cortex (PFC) of treated mice, a most involved area of the brain in schizophrenia. We show that intranasal delivery of MSC-EVs improve social interaction and disruption in prepulse inhibition (PPI) seen in PCP-treated mice. In addition, immunohistochemical studies demonstrate that the EVs preserve the number of parvalbumin-positive GABAergic interneurons in the PFC of treated mice. Finally, MSCs-EVs reduced glutamate levels in the CSF of PCP-treated mice, which might explain the reduction of toxicity. In conclusion, we show that MSCs-EVs improve the core schizophrenia-like behavior and biochemical markers of schizophrenia and might be used as a novel treatment for this incurable disorder.


Subject(s)
Extracellular Vesicles , Mesenchymal Stem Cells , Schizophrenia , Animals , Disease Models, Animal , Humans , Mice , Phencyclidine , Prefrontal Cortex , Schizophrenia/therapy
7.
Transl Psychiatry ; 10(1): 327, 2020 Sep 22.
Article in English | MEDLINE | ID: mdl-32963225

ABSTRACT

In the original Article, Dr. Angela Ruban's name was misspelled as "Aangela Ruban". This has been corrected in the PDF, HTML, and XML versions of this Article.

8.
Cell Death Dis ; 11(7): 578, 2020 07 24.
Article in English | MEDLINE | ID: mdl-32709881

ABSTRACT

APOE4 is a major risk factor for sporadic Alzheimer's disease; however, it is unclear how it exerts its pathological effects. Others and we have previously shown that autophagy is impaired in APOE4 compared to APOE3 astrocytes, and demonstrated differences in the expression of mitochondrial dynamics proteins in brains of APOE3 and APOE4 transgenic mice. Here, we investigated the effect of APOE4 expression on several aspects of mitochondrial function and network dynamics, including fusion, fission, and mitophagy, specifically in astrocytes. We found that APOE3 and APOE4 astrocytes differ in their mitochondrial dynamics, suggesting that the mitochondria of APOE4 astrocytes exhibit reduced fission and mitophagy. APOE4 astrocytes also show impaired mitochondrial function. Importantly, the autophagy inducer rapamycin enhanced mitophagy and improved mitochondrial functioning in APOE4 astrocytes. Collectively, the results demonstrate that APOE4 expression is associated with altered mitochondrial dynamics, which might lead to impaired mitochondrial function in astrocytes. This, in turn, may contribute to the pathological effects of APOE4 in Alzheimer's disease.


Subject(s)
Apolipoprotein E4/metabolism , Astrocytes/metabolism , Mitochondrial Dynamics , Apolipoprotein E3/metabolism , Astrocytes/ultrastructure , Carbonyl Cyanide m-Chlorophenyl Hydrazone/pharmacology , Cell Line , Humans , Lysosomes/drug effects , Lysosomes/metabolism , Mitochondria/metabolism , Mitochondria/ultrastructure , Mitochondrial Proteins/metabolism , Proteasome Endopeptidase Complex/drug effects , Proteasome Endopeptidase Complex/metabolism , Proteolysis/drug effects , Sirolimus/pharmacology , Ubiquitination/drug effects
9.
J Neurosurg Spine ; : 1-13, 2020 Jul 03.
Article in English | MEDLINE | ID: mdl-32619986

ABSTRACT

OBJECTIVE: Excitotoxicity due to neuronal damage and glutamate release is one of the first events that leads to the progression of neuronal degeneration and functional impairment. This study is based on a paradigm shift in the therapeutic approach for treating spinal cord injury (SCI). The authors tested a new treatment targeting removal of CNS glutamate into the blood circulation by injection of the blood glutamate scavengers (BGSs) recombinant enzyme glutamate-oxaloacetate transaminase (rGOT1) and its cosubstrate oxaloacetic acid (OxAc). Their primary objective was to investigate whether BGS treatment, followed by treadmill exercises in mice with SCI, could attenuate excitotoxicity, inflammation, scarring, and axonal degeneration and, at a later time point, improve functional recovery. METHODS: A pharmacokinetic experiment was done in C57BL/6 naive mice to verify rGOT1/OxAc blood activity and to characterize the time curve of glutamate reduction in the blood up to 24 hours. The reduction of glutamate in CSF after BGS administration in mice with SCI was confirmed by high-performance liquid chromatography. Next, SCI (left hemisection) was induced in the mice, and the mice were randomly assigned to one of the following groups at 1 hour postinjury: control (underwent SCI and received PBS), treadmill exercises, rGOT1/OxAc treatment, or rGOT1/OxAc treatment followed by treadmill exercises. Treatment started 1 hour postinjury with an injection of rGOT1/OxAc and continued for 5 consecutive days. Starting 1 week after SCI, the exercises and the combined treatment groups recommenced the treadmill exercise regimen 5 days a week for 3 months. Locomotor function was assessed for 3 months using the horizontal grid walking test and CatWalk. Axonal anterograde and wallerian degenerations were evaluated using tetramethylrhodamine dextran. Tissue sections were immunofluorescently stained for Iba1, GFAP, GAP-43, synaptophysin, and NeuN. RESULTS: BGS treatment decreased the CSF glutamate level up to 50%, reduced axonal wallerian degeneration, and increased axonal survival and GAP-43 expression in neuronal cells. Combined treatment reduced inflammation, scarring, and lesion size. Additionally, the combination of BGS treatment and exercises increased synapses around motor neurons and enhanced axonal regeneration through the lesion site. This resulted in motor function improvement 3 months post-SCI. CONCLUSIONS: As shown by biochemical, immunohistochemical, and functional analysis, BGSs exhibit a substantial neuroprotective effect by reducing excitotoxicity and secondary damage after SCI. Furthermore, in combination with exercises, they reduced axonal degeneration and scarring and resulted in improved functional recovery.

10.
Neural Dev ; 13(1): 24, 2018 11 17.
Article in English | MEDLINE | ID: mdl-30447699

ABSTRACT

BACKGROUND: Despite conserved developmental processes and organization of the vertebrate central nervous system, only some vertebrates including zebrafish can efficiently regenerate neural damage including after spinal cord injury. The mammalian spinal cord shows very limited regeneration and neurogenesis, resulting in permanent life-long functional impairment. Therefore, there is an urgent need to identify the cellular and molecular mechanisms that can drive efficient vertebrate neurogenesis following injury. A key pathway implicated in zebrafish neurogenesis is fibroblast growth factor signaling. METHODS: In the present study we investigated the roles of distinct fibroblast growth factor members and their receptors in facilitating different aspects of neural development and regeneration at different timepoints following spinal cord injury. After spinal cord injury in adults and during larval development, loss and/or gain of Fgf signaling was combined with immunohistochemistry, in situ hybridization and transgenes marking motor neuron populations in in vivo zebrafish and in vitro mammalian PC12 cell culture models. RESULTS: Fgf3 drives neurogenesis of Islet1 expressing motor neuron subtypes and mediate axonogenesis in cMet expressing motor neuron subtypes. We also demonstrate that the role of Fgf members are not necessarily simple recapitulating development. During development Fgf2, Fgf3 and Fgf8 mediate neurogenesis of Islet1 expressing neurons and neuronal sprouting of both, Islet1 and cMet expressing motor neurons. Strikingly in mammalian PC12 cells, all three Fgfs increased cell proliferation, however, only Fgf2 and to some extent Fgf8, but not Fgf3 facilitated neurite outgrowth. CONCLUSIONS: This study demonstrates differential Fgf member roles during neural development and adult regeneration, including in driving neural proliferation and neurite outgrowth of distinct spinal cord neuron populations, suggesting that factors including Fgf type, age of the organism, timing of expression, requirements for different neuronal populations could be tailored to best drive all of the required regenerative processes.


Subject(s)
Fibroblast Growth Factors/metabolism , Nerve Regeneration/physiology , Neurogenesis/physiology , Spinal Cord Injuries/metabolism , Animals , Animals, Genetically Modified , Cell Proliferation , Motor Neurons/metabolism , Neuroglia/cytology , Neuroglia/metabolism , Signal Transduction/physiology , Spinal Cord/cytology , Spinal Cord/metabolism , Zebrafish , Zebrafish Proteins/metabolism
11.
J Neurotrauma ; 35(21): 2581-2590, 2018 11 01.
Article in English | MEDLINE | ID: mdl-29495919

ABSTRACT

Neurotrauma causes immediate elevation of extracellular glutamate (Glu) levels, which creates excitotoxicity and facilitates inflammation, glial scar formation, and consequently neuronal death. Finding factors that reduce the inflammatory response and glial scar formation, and increase neuronal survival and neurite outgrowth, are of major importance for improving the outcome after spinal cord injury (SCI). In the present study, we evaluated a new treatment aiming to remove central nervous system (CNS) Glu into the systemic blood circulation by intravenous (IV) administration of blood Glu scavengers (BGS) such as the enzyme recombinant glutamate-oxaloacetate transaminase 1 (rGOT1) and its co-substrate. In this study we induced in mice an SCI (hemisection), and 1 h post-injury started administering BGS treatment for 5 consecutive days. The treatment reduced the expression levels of p-p38, which regulates apoptosis and increased the expression of p-Akt, which mediates cell survival. Moreover, this treatment decreased pro-inflammatory cytokine expression and microglia activation, reduced astrocytes' reactivity, and facilitated expression of radial glia markers such as Pax6 and nestin. BGS treatment increased the survival of neurons at lesion site and enabled axonal regeneration into the injury site. These effects were correlated with improved functional recovery of the left paretic hindlimb. Thus, early pharmacological intervention with BGS following SCI may be neuroprotective and create a pro-regenerative environment by modulating glia cell response. In light of our results, the availability of the method to remove excess Glu from CNS without the need to deliver drugs across the blood-brain barrier (BBB) and with minimal or no adverse effects may provide a major therapeutic asset.


Subject(s)
Aspartate Aminotransferase, Cytoplasmic/pharmacology , Glutamic Acid/blood , Glutamic Acid/drug effects , Neuroprotective Agents/pharmacology , Spinal Cord Injuries/blood , Animals , Female , Male , Mice , Mice, Inbred C57BL , Recombinant Proteins/pharmacology
12.
Cell Death Dis ; 9(2): 47, 2018 01 19.
Article in English | MEDLINE | ID: mdl-29352243

ABSTRACT

ErbB2, a member of the ErbB family of receptor tyrosine kinases, is an essential player in the cell's growth and proliferation signaling pathways. Amplification or overexpression of ErbB2 is observed in ∼30% of breast cancer patients, and often drives cellular transformation and cancer development. Recently, we have shown that ErbB2 interacts with the nuclear-cytoplasmic shuttling protein nucleolin, an interaction which enhances cell transformation in vitro, and increases mortality risk and disease progression rate in human breast cancer patients. Given these results, and since acquired resistance to anti-ErbB2-targeted therapy is a major obstacle in treatment of breast cancer, we have examined the therapeutic potential of targeting the ErbB2-nucleolin complex. The effect of the nucleolin-specific inhibitor GroA (AS1411) on ErbB2-positive breast cancer was tested in vivo, in a mouse xenograft model for breast cancer; as well as in vitro, alone and in combination with the ErbB2 kinase-inhibitor tyrphostin AG-825. Here, we show that in vivo treatment of ErbB2-positive breast tumor xenografts with GroA reduces tumor size and leads to decreased ErbB2-mediated signaling. Moreover, we found that co-treatment of breast cancer cell lines with GroA and the ErbB2 kinase-inhibitor tyrphostin AG-825 enhances the anti-cancer effects exerted by GroA alone in terms of cell viability, mortality, migration, and invasiveness. We, therefore, suggest a novel therapeutic approach, consisting of combined inhibition of ErbB2 and nucleolin, which has the potential to improve breast cancer treatment efficacy.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Benzothiazoles/pharmacology , Breast Neoplasms/drug therapy , Oligodeoxyribonucleotides/pharmacology , Phosphoproteins/antagonists & inhibitors , RNA-Binding Proteins/antagonists & inhibitors , Receptor, ErbB-2/antagonists & inhibitors , Tyrphostins/pharmacology , Animals , Aptamers, Nucleotide , Benzothiazoles/administration & dosage , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cell Line, Tumor , Disease Models, Animal , Drug Synergism , Female , Humans , MCF-7 Cells , Mice , Mice, Nude , Oligodeoxyribonucleotides/administration & dosage , Phosphoproteins/metabolism , RNA-Binding Proteins/metabolism , Receptor, ErbB-2/metabolism , Signal Transduction , Tyrphostins/administration & dosage , Xenograft Model Antitumor Assays , Nucleolin
13.
Sci Rep ; 6: 37678, 2016 11 24.
Article in English | MEDLINE | ID: mdl-27883058

ABSTRACT

Lysophosphatidic acid (LPA) is a unique bioactive lysophospholipid that induces pleiotropic effects in various cell types and organisms by acting on its specific receptors. LPA is mainly synthetised extracellularly by the ectonucleotide pyrophosphatase/phosphodiesterase 2/autotaxin (enpp2). Altered LPA signalling is associated with embryonic abnormalities, suggesting critical roles for LPA during development. However, the role of LPA signalling during early embryogenesis is not well established. We demonstrate that enpp2/LPA signalling in the early zebrafish embryo results in altered axis and midline formation, defects in left right (L-R) patterning, ciliogenesis of the Kupffer's vesicle (KV), through the modulation of cell migration during gastrulation in a lpar1-3 Rho/ROCK-dependant manner. Overall, this study demonstrates an essential role of enpp2/LPA signalling during early embryogenesis.


Subject(s)
Body Patterning , Embryo, Nonmammalian/enzymology , Phosphoric Diester Hydrolases/metabolism , Zebrafish Proteins/metabolism , Zebrafish/embryology , Zebrafish/metabolism , Animals , Cell Movement , Cilia/metabolism , Embryonic Development/genetics , Gene Expression Regulation, Developmental , Organogenesis , Phenotype , Phosphoric Diester Hydrolases/genetics , Zebrafish/genetics , Zebrafish Proteins/genetics , rho GTP-Binding Proteins/metabolism , rho-Associated Kinases/metabolism
14.
J Neurochem ; 138(6): 845-58, 2016 09.
Article in English | MEDLINE | ID: mdl-27399849

ABSTRACT

Glial scarring, formed by reactive astrocytes, is one of the major impediments for regeneration after spinal cord injury (SCI). Reactive astrocytes become hypertrophic, proliferate and secrete chondroitin sulphate proteoglycans into the extracellular matrix (ECM). Many studies have demonstrated that epidermal growth factor receptors (EGFR) can mediate astrocyte reactivity after neurotrauma. Previously we showed that there is crosstalk between nucleolin and EGFR that leads to increased EGFR activation followed by increased cell proliferation. Treatment with the nucleolin inhibitor GroA (AS1411) prevented these effects in vitro and in vivo. In this study, we hypothesized that similar interactions may mediate astrogliosis after SCI. Our results demonstrate that nucleolin and EGFR interaction may play a pivotal role in mediating astrocyte proliferation and reactivity after SCI. Moreover, we demonstrate that treatment with GroA reduces EGFR activation, astrocyte proliferation and chondroitin sulphate proteoglycans secretion, therefore promoting axonal regeneration and sprouting into the lesion site. Our results identify, for the first time, a role for the interaction between nucleolin and EGFR in astrocytes after SCI, indicating that nucleolin inhibitor GroA may be used as a novel treatment after neurotrauma. A major barrier for axonal regeneration after spinal cord injury is glial scar created by reactive and proliferating astrocytes. EGFR mediate astrocyte reactivity. We showed that inhibition of nucleolin by GroA, reduces EGFR activation, which results in attenuation of astrocyte reactivity and proliferation in vivo and in vitro. EGFR, epidermal growth factor receptor.


Subject(s)
ErbB Receptors/agonists , Neuroglia/pathology , Oligodeoxyribonucleotides/pharmacology , Phosphoproteins/antagonists & inhibitors , RNA-Binding Proteins/antagonists & inhibitors , Spinal Cord Injuries/pathology , Animals , Aptamers, Nucleotide , Astrocytes/drug effects , Behavior, Animal/drug effects , Cell Proliferation/drug effects , Chondroitin Sulfate Proteoglycans/metabolism , Female , Glial Fibrillary Acidic Protein/metabolism , Gliosis/pathology , Humans , Immunohistochemistry , Locomotion/genetics , Male , Mice , Mice, Inbred C57BL , Phosphoproteins/metabolism , RNA-Binding Proteins/metabolism , Spinal Cord Injuries/physiopathology , Spinal Cord Injuries/psychology , Nucleolin
15.
Mol Cell Neurosci ; 68: 82-91, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25936601

ABSTRACT

Spinal cord injury (SCI) frequently leads to a permanent functional impairment as a result of the initial injury followed by secondary injury mechanism, which is characterised by increased inflammation, glial scarring and neuronal cell death. Finding drugs that may reduce inflammatory cell invasion and activation to reduce glial scarring and increase neuronal survival is of major importance for improving the outcome after SCI. In the present study, we examined the effect of rapamycin, an mTORC1 inhibitor and an inducer of autophagy, on recovery from spinal cord injury. Autophagy, a process that facilitates the degradation of cytoplasmic proteins, is also important for maintenance of neuronal homeostasis and plays a major role in neurodegeneration after neurotrauma. We examined rapamycin effects on the inflammatory response, glial scar formation, neuronal survival and regeneration in vivo using spinal cord hemisection model in mice, and in vitro using primary cortical neurons and human astrocytes. We show that a single injection of rapamycin, inhibited p62/SQSTM1, a marker of autophagy, inhibited mTORC1 downstream effector p70S6K, reduced macrophage/neutrophil infiltration into the lesion site, microglia activation and secretion of TNFα. Rapamycin inhibited astrocyte proliferation and reduced the number of GFAP expressing cells at the lesion site. Finally, it increased neuronal survival and axonogenesis towards the lesion site. Our study shows that rapamycin treatment increased significantly p-Akt levels at the lesion site following SCI. Similarly, rapamycin treatment of neurons and astrocytes induced p-Akt elevation under stress conditions. Together, these findings indicate that rapamycin is a promising candidate for treatment of acute SCI condition and may be a useful therapeutic agent.


Subject(s)
Immunosuppressive Agents/therapeutic use , Inflammation/drug therapy , Inflammation/etiology , Sirolimus/therapeutic use , Spinal Cord Injuries/complications , Spinal Cord Injuries/pathology , Animals , Astrocytes/drug effects , Astrocytes/physiology , CD11b Antigen/metabolism , Cell Count , Cell Survival/drug effects , Cell Survival/physiology , Cells, Cultured , Disease Models, Animal , ELAV-Like Protein 3/metabolism , Gene Expression Regulation/drug effects , Glial Fibrillary Acidic Protein/metabolism , Humans , Ki-67 Antigen/metabolism , Male , Mice , Mice, Inbred C57BL , Neurons/drug effects , Rats , Time Factors
16.
Oncotarget ; 5(18): 8602-13, 2014 Sep 30.
Article in English | MEDLINE | ID: mdl-25261371

ABSTRACT

The three oncogenes, ErbB receptors, Ras proteins and nucleolin may contribute to malignant transformation. Previously, we demonstrated that nucleolin could bind both Ras protein and ErbB receptors. We also showed that the crosstalk between the three proteins facilitates anchorage independent growth and tumor growth in nude mice, and that inhibition of this interaction in prostate and colon cancer cells reduces tumorigenicity. In the present study, we show that treatment with Ras and nucleolin inhibitors reduces the oncogenic effect induced by ErbB1 receptor in U87-MG cells. This combined treatment enhances cell death, reduces cell proliferation and cell migration. Moreover, we demonstrate a pivotal role of nucleolin in ErbB1 activation by its ligand. Nucleolin inhibitor prevents EGF-induced receptor activation and its downstream signaling followed by reduced proliferation. Furthermore, inhibition of Ras by Salirasib (FTS), mainly reduces cell viability and motility. The combined treatment, which targets both Ras and nucleolin, additively reduces tumorigenicity both in vitro and in vivo. These results suggest that targeting both nucleolin and Ras may represent an additional opportunity for inhibiting cancers, including glioblastoma, that are driven by these oncogenes.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Brain Neoplasms/drug therapy , ErbB Receptors/metabolism , Glioblastoma/drug therapy , Phosphoproteins/antagonists & inhibitors , RNA-Binding Proteins/antagonists & inhibitors , ras Proteins/antagonists & inhibitors , Animals , Aptamers, Nucleotide , Brain Neoplasms/enzymology , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Cell Death/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Farnesol/administration & dosage , Farnesol/analogs & derivatives , Glioblastoma/genetics , Glioblastoma/metabolism , Glioblastoma/pathology , Humans , Mice, Nude , Oligodeoxyribonucleotides/administration & dosage , Phosphoproteins/genetics , Phosphoproteins/metabolism , Phosphorylation , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Receptor Cross-Talk , Salicylates/administration & dosage , Signal Transduction/drug effects , Time Factors , Tumor Burden/drug effects , ras Proteins/metabolism , Nucleolin
17.
Brain Behav ; 4(2): 187-200, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24683512

ABSTRACT

OBJECTIVES: A major impediment for recovery after mammalian spinal cord injury (SCI) is the glial scar formed by proliferating reactive astrocytes. Finding factors that may reduce glial scarring, increase neuronal survival, and promote neurite outgrowth are of major importance for improving the outcome after SCI. Exogenous fibroblast growth factor (Fgf) has been shown to decrease injury volume and improve functional outcome; however, the mechanisms by which this is mediated are still largely unknown. METHODS: In this study, Fgf2 was administered for 2 weeks in mice subcutaneously, starting 30 min after spinal cord hemisection. RESULTS: Fgf2 treatment decreased the expression of TNF-a at the lesion site, decreased monocyte/macrophage infiltration, and decreased gliosis. Fgf2 induced astrocytes to adopt a polarized morphology and increased expression of radial markers such as Pax6 and nestin. In addition, the levels of chondroitin sulfate proteoglycans (CSPGs), expressed by glia, were markedly decreased. Furthermore, Fgf2 treatment promotes the formation of parallel glial processes, "bridges," at the lesion site that enable regenerating axons through the injury site. Additionally, Fgf2 treatment increased Sox2-expressing cells in the gray matter and neurogenesis around and at the lesion site. Importantly, these effects were correlated with enhanced functional recovery of the left paretic hind limb. CONCLUSIONS: Thus, early pharmacological intervention with Fgf2 following SCI is neuroprotective and creates a proregenerative environment by the modulation of the glia response.


Subject(s)
Astrocytes/drug effects , Fibroblast Growth Factor 2/pharmacology , Gliosis/drug therapy , Neural Stem Cells/drug effects , Recovery of Function/drug effects , Spinal Cord Injuries/drug therapy , Spinal Cord Regeneration/drug effects , Animals , Male , Mice , Mice, Inbred C57BL
18.
Eur J Neurosci ; 39(9): 1419-28, 2014 May.
Article in English | MEDLINE | ID: mdl-24611983

ABSTRACT

Ephs form the largest family of receptor tyrosine kinases. They interact with the membrane-bound ligands - ephrins - to control crucial aspects of brain development. EphA4 is the most prominent member of the family in terms of versatility and ability to bind most ephrin ligands. EphA4 regulates brain development by modulating neuronal migration and connectivity. In the present study, we address the involvement of EphA4 in patterning the primary visual cortex (V1) of the marmoset monkey by characterizing the cellular expression profile of EphA4 from late embryonic stages to adulthood. We identified continuous expression on neurons in the cortical plate and mature neocortical layers, similar to that described in the mouse, excluding a role for EphA4 in the formation of borders between visual areas in the marmoset neocortex. In addition to neurons, we also report expression of EphA4 on glial populations, including radial glia and astrocytes. In contrast to what is seen in the mouse, EphA4 expression on astrocytes persists in the adult marmoset V1, including around blood vessels and in the white matter. Robust expression by glial populations, which retain neurogenic properties in the postnatal marmoset, indicates that EphA4 may have acquired additional roles during evolution, with important implications for the benefits of EphA4-blocking therapies following brain injury.


Subject(s)
Neuroglia/metabolism , Neurons/metabolism , Receptor, EphA4/metabolism , Visual Cortex/enzymology , Visual Cortex/growth & development , Animals , Callithrix , Female , Male , Mice
19.
J Neuroinflammation ; 11: 37, 2014 Feb 27.
Article in English | MEDLINE | ID: mdl-24576351

ABSTRACT

BACKGROUND: Lysophosphatidic acid (LPA) is a bioactive phospholipid with a potentially causative role in neurotrauma. Blocking LPA signaling with the LPA-directed monoclonal antibody B3/Lpathomab is neuroprotective in the mouse spinal cord following injury. FINDINGS: Here we investigated the use of this agent in treatment of secondary brain damage consequent to traumatic brain injury (TBI). LPA was elevated in cerebrospinal fluid (CSF) of patients with TBI compared to controls. LPA levels were also elevated in a mouse controlled cortical impact (CCI) model of TBI and B3 significantly reduced lesion volume by both histological and MRI assessments. Diminished tissue damage coincided with lower brain IL-6 levels and improvement in functional outcomes. CONCLUSIONS: This study presents a novel therapeutic approach for the treatment of TBI by blocking extracellular LPA signaling to minimize secondary brain damage and neurological dysfunction.


Subject(s)
Brain Injuries/drug therapy , Brain Injuries/immunology , Immunoglobulin G/therapeutic use , Immunologic Factors/therapeutic use , Lysophospholipids/immunology , Adult , Aged, 80 and over , Animals , Brain Injuries/cerebrospinal fluid , Cytokines/metabolism , Disease Models, Animal , Female , Glasgow Coma Scale , Humans , Lysophospholipids/cerebrospinal fluid , Male , Mice , Mice, Inbred C57BL , Middle Aged , Single-Blind Method , Young Adult
20.
J Lipid Res ; 54(5): 1192-206, 2013 May.
Article in English | MEDLINE | ID: mdl-23463731

ABSTRACT

We previously reported that lysophosphatidic acid (LPA) inhibits the neuronal differentiation of human embryonic stem cells (hESC). We extended these studies by analyzing LPA's effects on the expansion of neural stem/progenitor cells (NS/PC) derived from hESCs and human induced pluripotent stem cells (iPSC), and we assessed whether data obtained on the neural differentiation of hESCs were relevant to iPSCs. We showed that hESCs and iPSCs exhibited comparable mRNA expression profiles of LPA receptors and producing enzymes upon neural differentiation. We demonstrated that LPA inhibited the expansion of NS/PCs of both origins, mainly by increased apoptosis in a Rho/Rho-associated kinase (ROCK)-dependent mechanism. Furthermore, LPA inhibited the neuronal differentiation of iPSCs. Lastly, LPA induced neurite retraction of NS/PC-derived early neurons through Rho/ROCK, which was accompanied by myosin light chain (MLC) phosphorylation. Our data demonstrate the consistency of LPA effects across various sources of human NS/PCs, rendering hESCs and iPSCs valuable models for studying lysophospholipid signaling in human neural cells. Our data also highlight the importance of the Rho/ROCK pathway in human NS/PCs. As LPA levels are increased in the central nervous system (CNS) following injury, LPA-mediated effects on NS/PCs and early neurons could contribute to the poor neurogenesis observed in the CNS following injury.


Subject(s)
Lysophospholipids/pharmacology , Neurons/cytology , rho GTP-Binding Proteins/metabolism , rho-Associated Kinases/metabolism , Cell Differentiation , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/drug effects , Induced Pluripotent Stem Cells/metabolism , Myosin Light Chains/metabolism , Neural Stem Cells/cytology , Neural Stem Cells/drug effects , Neural Stem Cells/metabolism , Neurons/metabolism , Phosphorylation , Receptors, Lysophosphatidic Acid/metabolism , Signal Transduction/drug effects , rho GTP-Binding Proteins/genetics , rho-Associated Kinases/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...