Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
iScience ; 25(8): 104813, 2022 Aug 19.
Article in English | MEDLINE | ID: mdl-35982785

ABSTRACT

Species differences in brain and blood-brain barrier (BBB) biology hamper the translation of findings from animal models to humans, impeding the development of therapeutics for brain diseases. Here, we present a human organotypic microphysiological system (MPS) that includes endothelial-like cells, pericytes, glia, and cortical neurons and maintains BBB permeability at in vivo relevant levels. This human Brain-Chip engineered to recapitulate critical aspects of the complex interactions that mediate neuroinflammation and demonstrates significant improvements in clinical mimicry compared to previously reported similar MPS. In comparison to Transwell culture, the transcriptomic profiling of the Brain-Chip displayed significantly advanced similarity to the human adult cortex and enrichment in key neurobiological pathways. Exposure to TNF-α recreated the anticipated inflammatory environment shown by glia activation, increased release of proinflammatory cytokines, and compromised barrier permeability. We report the development of a robust brain MPS for mechanistic understanding of cell-cell interactions and BBB function during neuroinflammation.

2.
ALTEX ; 39(4): 694-709, 2022.
Article in English | MEDLINE | ID: mdl-35404466

ABSTRACT

Parkinson's disease (PD) is a complex neurodegenerative condition with a multifactorial origin. To date, approaches to drug discovery for PD have resulted in symptomatic therapies for the motor manifestations and signs associated with neurodegeneration but have failed to identify preventive or curative therapies. This failure mainly originates from the persistence of major gaps in our understanding of the specific molecular basis of PD initiation and progression. New approach methodologies (NAMs) hold the potential to advance PD research while facilitating a move away from ani-mal-based research. We report a workshop involving NAM experts in the field of PD and neurodegenerative diseases, who discussed and identified a scientific strategy for successful, human-specific PD research. We outline some of the most important human-specific NAMs, along with their main potentials and limitations, and suggest possible ways to overcome the latter. Key recommendations to advance PD research include integrating NAMs while accounting for multiple levels of complexity, from molecular to population level; learning from recent advances in Alzheimer's disease research; increasing the sharing of data; promoting innovative pilot studies on disease pathogenesis; and accessing philanthropic funding to enable studies using novel approaches. Collaborative efforts between different stakeholders, including researchers, clinicians, and funding agencies, are urgently needed to create a scientific roadmap and support a paradigm change towards effective, human-specific research for neurodegenerative diseases without animals, as is already happening in the field of toxicology.


Subject(s)
Parkinson Disease , Animals , Humans , Parkinson Disease/diagnosis , Parkinson Disease/drug therapy , Drug Discovery
3.
Nat Commun ; 12(1): 5907, 2021 10 08.
Article in English | MEDLINE | ID: mdl-34625559

ABSTRACT

Parkinson's disease and related synucleinopathies are characterized by the abnormal accumulation of alpha-synuclein aggregates, loss of dopaminergic neurons, and gliosis of the substantia nigra. Although clinical evidence and in vitro studies indicate disruption of the Blood-Brain Barrier in Parkinson's disease, the mechanisms mediating the endothelial dysfunction is not well understood. Here we leveraged the Organs-on-Chips technology to develop a human Brain-Chip representative of the substantia nigra area of the brain containing dopaminergic neurons, astrocytes, microglia, pericytes, and microvascular brain endothelial cells, cultured under fluid flow. Our αSyn fibril-induced model was capable of reproducing several key aspects of Parkinson's disease, including accumulation of phosphorylated αSyn (pSer129-αSyn), mitochondrial impairment, neuroinflammation, and compromised barrier function. This model may enable research into the dynamics of cell-cell interactions in human synucleinopathies and serve as a testing platform for target identification and validation of novel therapeutics.


Subject(s)
Blood-Brain Barrier/metabolism , Brain/metabolism , Parkinson Disease/metabolism , Synucleinopathies/metabolism , alpha-Synuclein/metabolism , Astrocytes/metabolism , Brain/diagnostic imaging , Brain/pathology , Dopaminergic Neurons/metabolism , Endothelial Cells/metabolism , Gliosis/pathology , Humans , Microglia/metabolism , Mitochondria/metabolism , Pericytes/metabolism , Phosphorylation , Substantia Nigra/metabolism , Transcriptome
4.
Neuropharmacology ; 121: 140-157, 2017 Jul 15.
Article in English | MEDLINE | ID: mdl-28461162

ABSTRACT

Neurotrophic factors are among the most promising treatments aiming at slowing or stopping and even reversing Parkinson's disease (PD). However, in most cases, they cannot readily cross the human blood-brain-barrier (BBB). Herein, we propose as a therapeutic for PD the small molecule 17-beta-spiro-[5-androsten-17,2'-oxiran]-3beta-ol (BNN-20), a synthetic analogue of DHEA, which crosses the BBB and is deprived of endocrine side-effects. Using the "weaver" mouse, a genetic model of PD, which exhibits progressive dopaminergic neurodegeneration in the Substantia Nigra (SN), we have shown that long-term administration (P1-P21) of BNN-20 almost fully protected the dopaminergic neurons and their terminals, via i) a strong anti-apoptotic effect, probably mediated through the Tropomyosin receptor kinase B (TrkB) neurotrophin receptor's PI3K-Akt-NF-κB signaling pathway, ii) by exerting an efficient antioxidant effect, iii) by inducing significant anti-inflammatory activity and iv) by restoring Brain-Derived Neurotrophic Factor (BDNF) levels. By intercrossing "weaver" with NGL mice (dual GFP/luciferase-NF-κΒ reporter mice, NF-κΒ.GFP.Luc), we obtained Weaver/NGL mice that express the NF-κB reporter in all somatic cells. Acute BNN-20 administration to Weaver/NGL mice induced a strong NF-κB-dependent transcriptional response in the brain as detected by bioluminescence imaging, which was abolished by co-administration of the TrkB inhibitor ANA-12. This indicates that BNN-20 exerts its beneficial action (at least in part) through the TrkB-PI3K-Akt-NF-κB signaling pathway. These results could be of clinical relevance, as they suggest BNN-20 as an important neuroprotective agent acting through the TrkB neurotrophin receptor pathway, mimicking the action of the endogenous neurotrophin BDNF. Thus BNN-20 could be proposed for treatment of PD.


Subject(s)
Dehydroepiandrosterone/analogs & derivatives , Dopamine/metabolism , Dopaminergic Neurons/drug effects , Mesencephalon/cytology , Receptor, trkB/metabolism , Adjuvants, Immunologic/pharmacology , Animals , Animals, Newborn , Antigens, CD1/metabolism , Azepines/pharmacology , Benzamides/pharmacology , CHO Cells , Cricetulus , Dehydroepiandrosterone/pharmacology , Dopamine Plasma Membrane Transport Proteins/metabolism , Female , Male , Mesencephalon/drug effects , Mesencephalon/metabolism , Mice , Mice, Neurologic Mutants , Models, Genetic , Signal Transduction/drug effects , Signal Transduction/physiology , Tubulin/metabolism , Tyrosine 3-Monooxygenase/metabolism
5.
Sci Rep ; 7: 44418, 2017 03 14.
Article in English | MEDLINE | ID: mdl-28290516

ABSTRACT

Accumulating evidence during the last decades revealed that androgen can exert membrane initiated actions that involve signaling via specific kinases and the modulation of significant cellular processes, important for prostate cancer cell growth and metastasis. Results of the present work clearly show that androgens can specifically act at the membrane level via the GPCR oxoeicosanoid receptor 1 (OXER1) in prostate cancer cells. In fact, OXER1 expression parallels that of membrane androgen binding in prostate cancer cell lines and tumor specimens, while in silico docking simulation of OXER1 showed that testosterone could bind to OXER1 within the same grove as 5-OxoETE, the natural ligand of OXER1. Interestingly, testosterone antagonizes the effects of 5-oxoETE on specific signaling pathways and rapid effects such as actin cytoskeleton reorganization that ultimately can modulate cell migration and metastasis. These findings verify that membrane-acting androgens exert specific effects through an antagonistic interaction with OXER1. Additionally, this interaction between androgen and OXER1, which is an arachidonic acid metabolite receptor expressed in prostate cancer, provides a novel link between steroid and lipid actions and renders OXER1 as new player in the disease. These findings should be taken into account in the design of novel therapeutic approaches in prostate cancer.


Subject(s)
Androgens/chemistry , Cell Proliferation/genetics , Prostatic Neoplasms/genetics , Receptors, Eicosanoid/chemistry , Androgens/genetics , Arachidonic Acid/metabolism , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Humans , Male , Molecular Docking Simulation , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Receptors, Eicosanoid/genetics , Testosterone/chemistry , Testosterone/genetics
7.
Neuropharmacology ; 111: 266-282, 2016 12.
Article in English | MEDLINE | ID: mdl-27618740

ABSTRACT

Nerve growth factor (NGF) holds a pivotal role in brain development and maintenance, been also involved in the pathophysiology of neurodegenerative diseases. Here, we provide evidence that a novel C17-spiroepoxy steroid derivative, BNN27, specifically interacts with and activates the TrkA receptor of NGF, inducing phosphorylation of TrkA tyrosine residues and down-stream neuronal survival-related kinase signaling. Additionally, BNN27 potentiates the efficacy of low levels of NGF, by facilitating its binding to the TrkA receptors and differentially inducing fast return of internalized TrkA receptors into neuronal cell membranes. Furthermore, BNN27 synergizes with NGF in promoting axonal outgrowth, effectively rescues from apoptosis NGF-dependent and TrkA positive sympathetic and sensory neurons, in vitro, ex vivo and in vivo in NGF null mice. Interestingly, BNN27 does not possess the hyperalgesic properties of NGF. BNN27 represents a lead molecule for the development of neuroprotective TrkA receptor agonists, with potential therapeutic applications in neurodegenerative diseases and in brain trauma.


Subject(s)
Cell Differentiation/drug effects , Cell Survival/drug effects , Dehydroepiandrosterone/pharmacology , Nerve Growth Factor/pharmacology , Neurons/drug effects , Neurons/metabolism , Receptor, trkA/metabolism , Animals , Apoptosis/drug effects , Axons/drug effects , Axons/metabolism , Binding Sites , CHO Cells , Cricetulus , Dehydroepiandrosterone/chemistry , HEK293 Cells , Humans , Hyperalgesia/chemically induced , Mice , Mice, Knockout , Models, Molecular , Molecular Dynamics Simulation , Nerve Growth Factor/genetics , Neuroglia/drug effects , Neuroglia/metabolism , PC12 Cells , Phosphorylation , Rats , Receptor, trkA/agonists , Recombinant Proteins/pharmacology , Sensory Receptor Cells/drug effects , Sensory Receptor Cells/metabolism , Signal Transduction
8.
Front Pharmacol ; 7: 512, 2016.
Article in English | MEDLINE | ID: mdl-28082899

ABSTRACT

Neurotrophin receptors mediate a plethora of signals affecting neuronal survival. The p75 pan-neurotrophin receptor controls neuronal cell fate after its selective activation by immature and mature isoforms of all neurotrophins. It also exerts pleiotropic effects interacting with a variety of ligands in different neuronal or non-neuronal cells. In the present study, we explored the biophysical and functional interactions of a blood-brain-barrier (BBB) permeable, C17-spiroepoxy steroid derivative, BNN27, with p75NTR receptor. BNN27 was recently shown to bind to NGF high-affinity receptor, TrkA. We now tested the p75NTR-mediated effects of BNN27 in mouse Cerebellar Granule Neurons (CGNs), expressing p75NTR, but not TrkA receptors. Our findings show that BNN27 physically interacts with p75NTR receptors in specific amino-residues of its extracellular domain, inducing the recruitment of p75NTR receptor to its effector protein RIP2 and the simultaneous release of RhoGDI in primary neuronal cells. Activation of the p75NTR receptor by BNN27 reverses serum deprivation-induced apoptosis of CGNs resulting in the decrease of the phosphorylation of pro-apoptotic JNK kinase and of the cleavage of Caspase-3, effects completely abolished in CGNs, isolated from p75NTR null mice. In conclusion, BNN27 represents a lead molecule for the development of novel p75NTR ligands, controlling specific p75NTR-mediated signaling of neuronal cell fate, with potential applications in therapeutics of neurodegenerative diseases and brain trauma.

9.
Endocrinology ; 156(1): 16-23, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25330101

ABSTRACT

Dehydroepiandosterone (DHEA), the most abundant steroid in humans, affects multiple cellular functions of the endocrine, immune, and nervous systems. However, up to quite recently, no receptor has been described specifically for it, whereas most of its physiological actions have been attributed to its conversion to either androgens or estrogens. DHEA interacts and modulate a variety of membrane and intracellular neurotransmitter and steroid receptors. We have recently reported that DHEA protects neuronal cells against apoptosis, interacting with TrkA, the high-affinity prosurvival receptor of the neurotrophin, nerve growth factor. Intrigued by its pleiotropic effects in the nervous system of a variety of species, we have investigated the ability of DHEA to interact with the other two mammalian neurotrophin receptors, ie, the TrkB and TrkC, as well as their invertebrate counterparts (orthologs) in mollusks Lymnaea and Aplysia and in cephalochordate fish Amphioxus. Amazingly, DHEA binds to all Trk receptors, although with lower affinity by 2 orders of magnitude compared with that of the polypeptidic neurotrophins. DHEA effectively induced the first step of the TrkA and TrkC receptors activation (phosphorylation at tyrosine residues), including the vertebrate neurotrophin nonresponding invertebrate Lymnaea and Aplysia receptors. Based on our data, we hypothesize that early in evolution, DHEA may have acted as a nonspecific neurotrophic factor promoting neuronal survival. The interaction of DHEA with all types of neurotrophin receptors offers new insights into the largely unidentified mechanisms of its actions on multiple tissues and organs known to express neurotrophin receptors.


Subject(s)
Dehydroepiandrosterone/metabolism , Evolution, Molecular , Phylogeny , Receptors, Nerve Growth Factor/metabolism , Animals , Dehydroepiandrosterone/chemistry , Extracellular Signal-Regulated MAP Kinases/genetics , Extracellular Signal-Regulated MAP Kinases/metabolism , Gene Expression Regulation/physiology , Invertebrates , Ligands , Mammals , Molecular Structure , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism
10.
Endocrinology ; 154(7): 2446-56, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23696568

ABSTRACT

Tumor growth is fostered by inhibition of cell death, which involves the receptiveness of tumor to growth factors and hormones. We have recently shown that testosterone exerts proapoptotic effects in prostate and colon cancer cells through a membrane-initiated mechanism. In addition, we have recently reported that dehydroepiandrosterone (DHEA) can control cell fate, activating nerve growth factor (NGF) receptors, namely tropomyosin-related kinase (Trk)A and p75 neurotrophin receptor, in primary neurons and in PC12 tumoral cells. NGF was recently involved in cancer cell proliferation and apoptosis. In the present study, we explored the cross talk between androgens (testosterone and DHEA) and NGF in regulating apoptosis of prostate and colon cancer cells. DHEA and NGF strongly blunted serum deprivation-induced apoptosis, whereas testosterone induced apoptosis of both cancer cell lines. The antiapoptotic effect of both DHEA and NGF was completely reversed by testosterone. In line with this, DHEA or NGF up-regulated, whereas testosterone down-regulated, the expression of TrkA receptor. The effects of androgens were abolished in both cell lines in the presence of TrkA inhibitor. DHEA induced the phosphorylation of TrkA and the interaction of p75 neurotrophin receptor with its effectors, Rho protein GDP dissociation inhibitor and receptor interacting serine/threonine-protein kinase 2. Conversely, testosterone was unable to activate both receptors. Testosterone acted as a DHEA and NGF antagonist, by blocking the activation of both receptors by DHEA or NGF. Our findings suggest that androgens may influence hormone-sensitive tumor cells via their cross talk with NGF receptors. The interplay between steroid hormone and neurotrophins signaling in hormone-dependent tumors offers new insights in the pathophysiology of these neoplasias.


Subject(s)
Dehydroepiandrosterone/pharmacology , Receptors, Nerve Growth Factor/metabolism , Testosterone/pharmacology , Animals , Apoptosis/drug effects , Cell Proliferation/drug effects , Colonic Neoplasms/metabolism , Humans , Male , Nerve Growth Factor/pharmacology , Nerve Growth Factors/genetics , Nerve Growth Factors/metabolism , PC12 Cells , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Rats , Receptor, Nerve Growth Factor/genetics , Receptor, Nerve Growth Factor/metabolism , Receptors, Nerve Growth Factor/genetics , Signal Transduction/drug effects , Tumor Cells, Cultured
11.
Cell Rep ; 2(6): 1563-70, 2012 Dec 27.
Article in English | MEDLINE | ID: mdl-23260665

ABSTRACT

Structural determinants underlying signaling specificity in the tumor necrosis factor receptor superfamily (TNFRSF) are poorly characterized, and it is unclear whether different signaling outputs can be genetically dissociated. The p75 neurotrophin receptor (p75(NTR)), also known as TNFRSF16, is a key regulator of trophic and injury responses in the nervous system. Here, we describe a genetic approach for dissecting p75(NTR) signaling and deciphering its underlying logic. Structural determinants important for regulation of cell death, NF-κB, and RhoA pathways were identified in the p75(NTR) death domain (DD). Proapoptotic and prosurvival pathways mapped onto nonoverlapping epitopes, demonstrating that different signaling outputs can be genetically separated in p75(NTR). Dissociation of c-Jun kinase (JNK) and caspase-3 activities indicated that JNK is necessary but not sufficient for p75(NTR)-mediated cell death. RIP2 recruitment and RhoGDI release were mechanistically linked, indicating that competition for DD binding underlies crosstalk between NF-κB and RhoA pathways in p75(NTR) signaling. These results provide insights into the logic of p75(NTR) signaling and pave the way for a genetic dissection of p75(NTR) function and physiology.


Subject(s)
MAP Kinase Kinase 4/metabolism , MAP Kinase Signaling System/physiology , Nerve Growth Factors/metabolism , Nerve Tissue Proteins/metabolism , Receptors, Nerve Growth Factor/metabolism , Animals , Caspase 3/genetics , Caspase 3/metabolism , HEK293 Cells , Humans , MAP Kinase Kinase 4/genetics , Mice , Mice, Knockout , NF-kappa B/genetics , NF-kappa B/metabolism , NIH 3T3 Cells , Nerve Growth Factors/genetics , Nerve Tissue Proteins/genetics , Protein Structure, Tertiary , Rats , Receptor-Interacting Protein Serine-Threonine Kinase 2/genetics , Receptor-Interacting Protein Serine-Threonine Kinase 2/metabolism , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Receptors, Growth Factor , Receptors, Nerve Growth Factor/genetics , rho GTP-Binding Proteins/genetics , rho GTP-Binding Proteins/metabolism , rho Guanine Nucleotide Dissociation Inhibitor alpha/genetics , rho Guanine Nucleotide Dissociation Inhibitor alpha/metabolism , rhoA GTP-Binding Protein/genetics , rhoA GTP-Binding Protein/metabolism
12.
J Immunol ; 189(10): 4748-58, 2012 Nov 15.
Article in English | MEDLINE | ID: mdl-23071284

ABSTRACT

The TNF superfamily ligands APRIL and BAFF bind with different affinity to two receptors, BCMA and TACI, and induce cell survival and/or proliferation, whereas BAFF also binds specifically to BAFFR. These molecules were considered specific for the immune system. Recently, however, they were also found in epithelial and mesenchymal noncancerous and cancerous tissues and cell lines. In this article, we report that hepatocellular carcinoma (HCC) cell lines HepG2 and Hep3B and HCC specimens express APRIL and BAFF and their receptors BCMA and BAFFR, but not TACI; APRIL/BCMA is enhanced in HCC, compared with normal liver tissue. In contrast to previous reports, APRIL binding to BCMA decreases cell proliferation by inducing G(2)/M cell cycle arrest, whereas BAFF has no effect on cell growth. HCC cells therefore represent a rare system in which these two ligands (APRIL and BAFF) exert a differential effect and may serve as a model for specific APRIL/BCMA actions. We show that the effect of APRIL is mediated via BCMA, which does not activate the classical NF-κB pathway, whereas it induces a novel signaling pathway, which involves JNK2 phosphorylation, FOXO3A activation, and GADD45 transcription. In addition, JNK2 mediates the phosphorylation of Akt, which is activated but does not participate in the antiproliferative effect of APRIL. Furthermore, transcriptome analysis revealed that APRIL modifies genes specifically related to cell cycle modulation, including MCM2/4/5/6, CDC6, PCNA, and POLE2. Our data, therefore, identify a novel APRIL/BCMA signaling pathway in HCC and suggest that APRIL could have a pleiotropic role in tumor biology.


Subject(s)
B-Cell Maturation Antigen/immunology , Cell Cycle Proteins/immunology , DNA-Binding Proteins/immunology , Forkhead Transcription Factors/immunology , G2 Phase Cell Cycle Checkpoints/immunology , Liver/immunology , M Phase Cell Cycle Checkpoints/immunology , MAP Kinase Kinase 7/immunology , Nuclear Proteins/immunology , Transcription Factors/immunology , B-Cell Activating Factor/genetics , B-Cell Activating Factor/immunology , B-Cell Activating Factor/metabolism , B-Cell Maturation Antigen/genetics , B-Cell Maturation Antigen/metabolism , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Forkhead Box Protein O3 , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , G2 Phase Cell Cycle Checkpoints/genetics , Hep G2 Cells , Humans , Liver/cytology , M Phase Cell Cycle Checkpoints/genetics , MAP Kinase Kinase 7/genetics , MAP Kinase Kinase 7/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Phosphorylation/genetics , Phosphorylation/immunology , Proliferating Cell Nuclear Antigen/genetics , Proliferating Cell Nuclear Antigen/immunology , Proliferating Cell Nuclear Antigen/metabolism , Protein Binding , Transcription Factors/genetics , Transcription Factors/metabolism , Transcription, Genetic/genetics , Transcription, Genetic/immunology
13.
Neuropharmacology ; 62(5-6): 2106-17, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22269901

ABSTRACT

The aim of the present study was to investigate the neuroprotective properties of the endogenous neurosteroid dehydroepiandrosterone (DHEA) in an in vivo model of retinal excitotoxicity, and the involvement of Nerve Growth Factor (NGF) in its actions. Adult Sprague-Dawley rats (250-300 g) received intravitreally (RS)-alpha-Amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid hydrobromide (AMPA; 42 nmol/eye) alone or in combination with DHEA (10(-8), 10(-7), 10(-6) M), or PBS (50 mM, control group). To examine the involvement of NGF and its TrkA receptor in the pharmacological effects of DHEA, animals received AMPA and NGF (60 pg/eye) in the absence or presence of a TrkA receptor inhibitor (Calbiochem 648450, 10(-6) M) or AMPA, DHEA (10(-6) M) and TrkA receptor inhibitor (10(-6), 10(-5) M). Immunohistochemistry studies [choline acetyltransferase (ChAT), brain nitric oxide synthetase (bNOS), calbindin, and TUNEL] and fluorescence-activated cell sorting (FACS) were used to examine retinal cell loss and protection. TrkA receptor immunoreactivity (-IR) and colocalization studies with relevant markers were also performed. AMPA (42 nmol) treatment resulted in a loss of bNOS, ChAT and calbindin immunoreactivities 24 h after its administration. DHEA, administered intravitreally, protected the retina from excitotoxicity in a dose-dependent manner. This effect was mimicked by NGF, and reversed by the NGF TrkA receptor inhibitor. The TrkA receptor is expressed in ganglion cells of rat retina. TUNEL staining and FACS analysis substantiated the neuroprotective actions of DHEA. These results demonstrate for the first time that the neurosteroid DHEA, administered intravitreally, protects the retina from AMPA excitotoxicity. An NGF TrkA receptor mechanism appears to be involved in this neuroprotection.


Subject(s)
Dehydroepiandrosterone/pharmacology , Excitatory Amino Acid Agonists/pharmacology , Neuroprotective Agents/pharmacology , Receptor, trkA/metabolism , Retina/drug effects , alpha-Amino-3-hydroxy-5-methyl-4-isoxazolepropionic Acid/pharmacology , Animals , Calbindins , Female , Male , Rats , Rats, Sprague-Dawley , Retina/metabolism , S100 Calcium Binding Protein G/metabolism
14.
J Cell Biochem ; 112(12): 3786-96, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21826705

ABSTRACT

Recently, our knowledge on estrogen receptor alpha (ERα) functions and fate has progressed: ERα enters in repeated transcription-modulating cycles (nucleus/cytoplasm/membrane trafficking processes and proteasomal degradation) that are governed by specific protein-protein interactions. Receptor fragments, especially those resulting from the proteolysis of its ligand binding domain, as well as corresponding synthetic peptides, have been studied with respect to their estrogenic/antiestrogenic potency. A peptide, corresponding to the human ERα P(295) -T(311) sequence (ERα17p) has been shown to alter breast cancer cell fate, triggering proliferation, or apoptosis. The aim of this work was to explore the effect of ERα17p on breast cancer cell migration and actin cytoskeleton dynamics and further analyze the mechanism of its membrane action. We show that ERα17p increases (MCF-7 and SK-BR-3 cells) or decreases (T47D and MDA-MB-231 cells) migration of breast cancer cells, in an ERα-independent manner, by mechanism(s) depending on Rho/ROCK and PI3K/Akt signaling pathways. Moreover, the peptide enhances the association of both estrogens and androgens to membranes and modifies cell migration, induced by E(2) -BSA. Additionally, initial evidence of a possible agonistic action of the peptide on GPR30 is also provided. ERα17p can be considered as a cell migration-modulator and could therefore constitute a therapeutic challenge, even in anti-estrogen-resistant tumors.


Subject(s)
Actins/metabolism , Breast Neoplasms/pathology , Cytoskeleton/metabolism , Estrogen Receptor alpha/physiology , Neoplasm Metastasis/pathology , Biopolymers/metabolism , Breast Neoplasms/metabolism , Cell Line, Tumor , Estrogen Receptor alpha/chemistry , Estrogen Receptor alpha/metabolism , Female , Humans , Protein Binding , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction
15.
PLoS Biol ; 9(4): e1001051, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21541365

ABSTRACT

The neurosteroid dehydroepiandrosterone (DHEA), produced by neurons and glia, affects multiple processes in the brain, including neuronal survival and neurogenesis during development and in aging. We provide evidence that DHEA interacts with pro-survival TrkA and pro-death p75(NTR) membrane receptors of neurotrophin nerve growth factor (NGF), acting as a neurotrophic factor: (1) the anti-apoptotic effects of DHEA were reversed by siRNA against TrkA or by a specific TrkA inhibitor; (2) [(3)H]-DHEA binding assays showed that it bound to membranes isolated from HEK293 cells transfected with the cDNAs of TrkA and p75(NTR) receptors (K(D): 7.4 ± 1.75 nM and 5.6 ± 0.55 nM, respectively); (3) immobilized DHEA pulled down recombinant and naturally expressed TrkA and p75(NTR) receptors; (4) DHEA induced TrkA phosphorylation and NGF receptor-mediated signaling; Shc, Akt, and ERK1/2 kinases down-stream to TrkA receptors and TRAF6, RIP2, and RhoGDI interactors of p75(NTR) receptors; and (5) DHEA rescued from apoptosis TrkA receptor positive sensory neurons of dorsal root ganglia in NGF null embryos and compensated NGF in rescuing from apoptosis NGF receptor positive sympathetic neurons of embryonic superior cervical ganglia. Phylogenetic findings on the evolution of neurotrophins, their receptors, and CYP17, the enzyme responsible for DHEA biosynthesis, combined with our data support the hypothesis that DHEA served as a phylogenetically ancient neurotrophic factor.


Subject(s)
Apoptosis , Dehydroepiandrosterone/metabolism , Nerve Tissue Proteins/metabolism , Neurons/pathology , Receptor, trkA/metabolism , Receptors, Nerve Growth Factor/metabolism , Animals , HEK293 Cells , Humans , Mice , Mice, Inbred C57BL , Nerve Tissue Proteins/genetics , Neurogenesis , PC12 Cells , Phosphorylation , Phylogeny , Proto-Oncogene Proteins c-bcl-2/metabolism , RNA Interference , Rats , Receptor, trkA/genetics , Receptors, Nerve Growth Factor/genetics , Signal Transduction , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...