29 results on '"Sclip A"'
Search Results
2. Combinatorial expression of neurexins and LAR-type phosphotyrosine phosphatase receptors instructs assembly of a cerebellar circuit.
- Author
-
Sclip A and Südhof TC
- Subjects
- Animals, Mice, Brain, Cognition, Mice, Knockout, Phosphotyrosine, Protein Tyrosine Phosphatases, Receptor-Like Protein Tyrosine Phosphatases, Class 2 genetics, Cerebellum, Purkinje Cells
- Abstract
Synaptic adhesion molecules (SAMs) shape the structural and functional properties of synapses and thereby control the information processing power of neural circuits. SAMs are broadly expressed in the brain, suggesting that they may instruct synapse formation and specification via a combinatorial logic. Here, we generate sextuple conditional knockout mice targeting all members of the two major families of presynaptic SAMs, Neurexins and leukocyte common antigen-related-type receptor phospho-tyrosine phosphatases (LAR-PTPRs), which together account for the majority of known trans-synaptic complexes. Using synapses formed by cerebellar Purkinje cells onto deep cerebellar nuclei as a model system, we confirm that Neurexins and LAR-PTPRs themselves are not essential for synapse assembly. The combinatorial deletion of both neurexins and LAR-PTPRs, however, decreases Purkinje-cell synapses on deep cerebellar nuclei, the major output pathway of cerebellar circuits. Consistent with this finding, combined but not separate deletions of neurexins and LAR-PTPRs impair motor behaviors. Thus, Neurexins and LAR-PTPRs are together required for the assembly of a functional cerebellar circuit., (© 2023. Springer Nature Limited.)
- Published
- 2023
- Full Text
- View/download PDF
3. Deletion of Calsyntenin-3, an atypical cadherin, suppresses inhibitory synapses but increases excitatory parallel-fiber synapses in cerebellum.
- Author
-
Liu Z, Jiang M, Liakath-Ali K, Sclip A, Ko J, Zhang RS, and Südhof TC
- Subjects
- Animals, Calcium-Binding Proteins, Cerebellum physiology, Membrane Proteins, Mice, Purkinje Cells physiology, Synaptic Transmission physiology, Cadherins, Synapses physiology
- Abstract
Cadherins contribute to the organization of nearly all tissues, but the functions of several evolutionarily conserved cadherins, including those of calsyntenins, remain enigmatic. Puzzlingly, two distinct, non-overlapping functions for calsyntenins were proposed: As postsynaptic neurexin ligands in synapse formation, or as presynaptic kinesin adaptors in vesicular transport. Here, we show that, surprisingly, acute CRISPR-mediated deletion of calsyntenin-3 in mouse cerebellum in vivo causes a large decrease in inhibitory synapse, but a robust increase in excitatory parallel-fiber synapses in Purkinje cells. As a result, inhibitory synaptic transmission was suppressed, whereas parallel-fiber synaptic transmission was enhanced in Purkinje cells by the calsyntenin-3 deletion. No changes in the dendritic architecture of Purkinje cells or in climbing-fiber synapses were detected. Sparse selective deletion of calsyntenin-3 only in Purkinje cells recapitulated the synaptic phenotype, indicating that calsyntenin-3 acts by a cell-autonomous postsynaptic mechanism in cerebellum. Thus, by inhibiting formation of excitatory parallel-fiber synapses and promoting formation of inhibitory synapses in the same neuron, calsyntenin-3 functions as a postsynaptic adhesion molecule that regulates the excitatory/inhibitory balance in Purkinje cells., Competing Interests: ZL, MJ, KL, AS, JK, RZ, TS No competing interests declared, (© 2022, Liu et al.)
- Published
- 2022
- Full Text
- View/download PDF
4. Neurexins regulate presynaptic GABA B -receptors at central synapses.
- Author
-
Luo F, Sclip A, Merrill S, and Südhof TC
- Subjects
- Animals, Brain Stem cytology, Brain Stem metabolism, CA1 Region, Hippocampal cytology, CA1 Region, Hippocampal physiology, CA3 Region, Hippocampal cytology, CA3 Region, Hippocampal metabolism, Calcium-Binding Proteins genetics, Cerebellum cytology, Cerebellum metabolism, Mice, Mice, Knockout, Models, Animal, Nerve Tissue Proteins genetics, Neural Cell Adhesion Molecules genetics, Neuronal Plasticity physiology, Patch-Clamp Techniques, Pyramidal Cells metabolism, Stereotaxic Techniques, Synaptic Transmission physiology, gamma-Aminobutyric Acid metabolism, Calcium-Binding Proteins metabolism, Nerve Tissue Proteins metabolism, Neural Cell Adhesion Molecules metabolism, Receptors, GABA-B metabolism, Synapses metabolism
- Abstract
Diverse signaling complexes are precisely assembled at the presynaptic active zone for dynamic modulation of synaptic transmission and synaptic plasticity. Presynaptic GABA
B -receptors nucleate critical signaling complexes regulating neurotransmitter release at most synapses. However, the molecular mechanisms underlying assembly of GABAB -receptor signaling complexes remain unclear. Here we show that neurexins are required for the localization and function of presynaptic GABAB -receptor signaling complexes. At four model synapses, excitatory calyx of Held synapses in the brainstem, excitatory and inhibitory synapses on hippocampal CA1-region pyramidal neurons, and inhibitory basket cell synapses in the cerebellum, deletion of neurexins rendered neurotransmitter release significantly less sensitive to GABAB -receptor activation. Moreover, deletion of neurexins caused a loss of GABAB -receptors from the presynaptic active zone of the calyx synapse. These findings extend the role of neurexins at the presynaptic active zone to enabling GABAB -receptor signaling, supporting the notion that neurexins function as central organizers of active zone signaling complexes.- Published
- 2021
- Full Text
- View/download PDF
5. Deorphanizing FAM19A proteins as pan-neurexin ligands with an unusual biosynthetic binding mechanism.
- Author
-
Khalaj AJ, Sterky FH, Sclip A, Schwenk J, Brunger AT, Fakler B, and Südhof TC
- Subjects
- Amino Acid Sequence, Animals, Cells, Cultured, Hippocampus metabolism, Ligands, Male, Mice, Mice, Inbred C57BL, Nerve Tissue Proteins metabolism, Synapses metabolism, Calcium-Binding Proteins metabolism, Chemokines metabolism, Neural Cell Adhesion Molecules metabolism, Neurons metabolism
- Abstract
Neurexins are presynaptic adhesion molecules that organize synapses by binding to diverse trans-synaptic ligands, but how neurexins are regulated is incompletely understood. Here we identify FAM19A/TAFA proteins, "orphan" cytokines, as neurexin regulators that interact with all neurexins, except for neurexin-1γ, via an unusual mechanism. Specifically, we show that FAM19A1-A4 bind to the cysteine-loop domain of neurexins by forming intermolecular disulfide bonds during transport through the secretory pathway. FAM19A-binding required both the cysteines of the cysteine-loop domain and an adjacent sequence of neurexins. Genetic deletion of neurexins suppressed FAM19A1 expression, demonstrating that FAM19As physiologically interact with neurexins. In hippocampal cultures, expression of exogenous FAM19A1 decreased neurexin O-glycosylation and suppressed its heparan sulfate modification, suggesting that FAM19As regulate the post-translational modification of neurexins. Given the selective expression of FAM19As in specific subtypes of neurons and their activity-dependent regulation, these results suggest that FAM19As serve as cell type-specific regulators of neurexin modifications., (© 2020 Khalaj et al.)
- Published
- 2020
- Full Text
- View/download PDF
6. Neurexins cluster Ca 2+ channels within the presynaptic active zone.
- Author
-
Luo F, Sclip A, Jiang M, and Südhof TC
- Subjects
- Animals, Exocytosis, Gene Deletion, Mice, Nerve Tissue Proteins metabolism, Synaptic Transmission, Calcium Channels metabolism, Nerve Tissue Proteins genetics, Synapses metabolism
- Abstract
To achieve ultrafast neurotransmission, neurons assemble synapses with highly organized presynaptic and postsynaptic nanomachines that are aligned by synaptic adhesion molecules. How functional assembly of presynaptic active zones is controlled via trans-synaptic interactions remains unknown. Here, we conditionally deleted all three neurexin adhesion molecules from presynaptic neurons of the calyx of Held in the mouse auditory system, a model synapse that allows precise biophysical analyses of synaptic properties. The pan-neurexin deletion had no effect on synapse development or the basic release machinery, but dramatically impaired fast neurotransmitter release. The overall properties of presynaptic calcium ion channels appeared normal, as reflected by the similar characteristics of calcium currents recorded at the nerve terminals. However, the pan-neurexin deletion significantly impaired the tight coupling of calcium influx to exocytosis, thereby suppressing neurotransmitter release. Furthermore, the pan-neurexin deletion reduced the function of calcium-activated BK potassium channels, whose activation depends on their tight association with presynaptic calcium channels. Together, these results suggest that neurexins perform a major function at the calyx synapse in coupling presynaptic calcium channels to release sites., (© 2020 The Authors.)
- Published
- 2020
- Full Text
- View/download PDF
7. LAR receptor phospho-tyrosine phosphatases regulate NMDA-receptor responses.
- Author
-
Sclip A and Südhof TC
- Subjects
- Animals, Female, Hippocampus metabolism, Hippocampus pathology, Male, Mice, Mice, Inbred C57BL, Mice, Knockout, Receptor-Like Protein Tyrosine Phosphatases, Class 2 genetics, Transcriptome, Neurons metabolism, Receptor-Like Protein Tyrosine Phosphatases, Class 2 metabolism, Receptors, N-Methyl-D-Aspartate metabolism, Synapses metabolism
- Abstract
LAR-type receptor phosphotyrosine-phosphatases (LAR-RPTPs) are presynaptic adhesion molecules that interact trans-synaptically with multitudinous postsynaptic adhesion molecules, including SliTrks, SALMs, and TrkC. Via these interactions, LAR-RPTPs are thought to function as synaptogenic wiring molecules that promote neural circuit formation by mediating the establishment of synapses. To test the synaptogenic functions of LAR-RPTPs, we conditionally deleted the genes encoding all three LAR-RPTPs, singly or in combination, in mice before synapse formation. Strikingly, deletion of LAR-RPTPs had no effect on synaptic connectivity in cultured neurons or in vivo, but impaired NMDA-receptor-mediated responses. Deletion of LAR-RPTPs decreased NMDA-receptor-mediated responses by a trans-synaptic mechanism. In cultured neurons, deletion of all LAR-RPTPs led to a reduction in synaptic NMDA-receptor EPSCs, without changing the subunit composition or the protein levels of NMDA-receptors. In vivo, deletion of all LAR-RPTPs in the hippocampus at birth also did not alter synaptic connectivity as measured via AMPA-receptor-mediated synaptic responses at Schaffer-collateral synapses monitored in juvenile mice, but again decreased NMDA-receptor mediated synaptic transmission. Thus, LAR-RPTPs are not essential for synapse formation, but control synapse properties by regulating postsynaptic NMDA-receptors via a trans-synaptic mechanism that likely involves binding to one or multiple postsynaptic ligands., Competing Interests: AS, TS No competing interests declared, (© 2020, Sclip and Südhof.)
- Published
- 2020
- Full Text
- View/download PDF
8. RIM-binding proteins recruit BK-channels to presynaptic release sites adjacent to voltage-gated Ca 2+ -channels.
- Author
-
Sclip A, Acuna C, Luo F, and Südhof TC
- Subjects
- Animals, Calcium Channels genetics, Large-Conductance Calcium-Activated Potassium Channels genetics, Mice, Synaptic Vesicles genetics, src Homology Domains, Calcium metabolism, Calcium Channels metabolism, Large-Conductance Calcium-Activated Potassium Channels metabolism, Presynaptic Terminals metabolism, Synaptic Vesicles metabolism
- Abstract
The active zone of presynaptic nerve terminals organizes the neurotransmitter release machinery, thereby enabling fast Ca
2+ -triggered synaptic vesicle exocytosis. BK-channels are Ca2+ -activated large-conductance K+ -channels that require close proximity to Ca2+ -channels for activation and control Ca2+ -triggered neurotransmitter release by accelerating membrane repolarization during action potential firing. How BK-channels are recruited to presynaptic Ca2+ -channels, however, is unknown. Here, we show that RBPs (for RIM-binding proteins), which are evolutionarily conserved active zone proteins containing SH3- and FN3-domains, directly bind to BK-channels. We find that RBPs interact with RIMs and Ca2+ -channels via their SH3-domains, but to BK-channels via their FN3-domains. Deletion of RBPs in calyx of Held synapses decreased and decelerated presynaptic BK-currents and depleted BK-channels from active zones. Our data suggest that RBPs recruit BK-channels into a RIM-based macromolecular active zone complex that includes Ca2+ -channels, synaptic vesicles, and the membrane fusion machinery, thereby enabling tight spatio-temporal coupling of Ca2+ -influx to Ca2+ -triggered neurotransmitter release in a presynaptic terminal., (© 2018 The Authors. Published under the terms of the CC BY 4.0 license.)- Published
- 2018
- Full Text
- View/download PDF
9. The c-jun N-terminal kinase plays a key role in ocular degenerative changes in a mouse model of Alzheimer disease suggesting a correlation between ocular and brain pathologies.
- Author
-
Buccarello L, Sclip A, Sacchi M, Castaldo AM, Bertani I, ReCecconi A, Maestroni S, Zerbini G, Nucci P, and Borsello T
- Abstract
Recently a range of ocular manifestations such as retinal and lens amyloid-beta accumulation and retinal nerve fiber layer loss have been proposed as potential biomarkers in Alzheimer disease (AD). The TgCRND8 mouse model of AD exhibits age-dependent amyloid β (Aβ) oligomers accumulation and cognitive defects, amyloid plaques and hyperphosphorylated Tau deposition and inflammation. We proved the correlation between ocular pathologies and AD, observing increased levels of p-APP and p-Tau, accumulation of Aβ oligomers in the retina, eye, and optic nerve. The accumulation of amyloid markers was significantly stronger in the retinal ganglion cell (RGC) layer, suggesting that RGC might be more susceptible to degeneration. We detected a thinning of the RGC layer as well as RGC death in the retina of TgCRND8 mice, by using a combination of Optical Coherence Tomography (OCT), immunofluorescence, immunohistochemistry and Western blotting techniques. We proved for the first time the key role of C-Jun N-terminal Kinase (JNK) in the ocular degeneration. In support of this, the administration of the JNK inhibitor, D-JNKI1, was able to counteract the Aβ and p-Tau accumulation in the retina of TgCRND8 mice, and consequently reduce RGCs loss. These results confirm that degenerative changes in the retina/eye of AD mouse model mirrors the events observed in the brain parenchyma. Ocular changes can be detected by non-invasive imaging techniques, such as OCT, to study and test different therapeutic strategies against degenerative events associated to AD., Competing Interests: CONFLICTS OF INTEREST The authors declare no actual or potential conflicts of interest.
- Published
- 2017
- Full Text
- View/download PDF
10. Evidence of Presynaptic Localization and Function of the c-Jun N-Terminal Kinase.
- Author
-
Biggi S, Buccarello L, Sclip A, Lippiello P, Tonna N, Rumio C, Di Marino D, Miniaci MC, and Borsello T
- Subjects
- Animals, Cerebral Cortex physiology, Excitatory Amino Acid Agonists pharmacology, Excitatory Postsynaptic Potentials, Female, Glycine pharmacology, Male, Mice, Mitogen-Activated Protein Kinase 10 metabolism, Mitogen-Activated Protein Kinase 9 metabolism, N-Methylaspartate pharmacology, Synaptosomes metabolism, Cerebral Cortex metabolism, JNK Mitogen-Activated Protein Kinases metabolism, Presynaptic Terminals enzymology, Receptors, N-Methyl-D-Aspartate metabolism, SNARE Proteins metabolism
- Abstract
The c-Jun N-terminal kinase (JNK) is part of a stress signalling pathway strongly activated by NMDA-stimulation and involved in synaptic plasticity. Many studies have been focused on the post-synaptic mechanism of JNK action, and less is known about JNK presynaptic localization and its physiological role at this site. Here we examined whether JNK is present at the presynaptic site and its activity after presynaptic NMDA receptors stimulation. By using N-SIM Structured Super Resolution Microscopy as well as biochemical approaches, we demonstrated that presynaptic fractions contained significant amount of JNK protein and its activated form. By means of modelling design, we found that JNK, via the JBD domain, acts as a physiological effector on T-SNARE proteins; then using biochemical approaches we demonstrated the interaction between Syntaxin-1-JNK, Syntaxin-2-JNK, and Snap25-JNK. In addition, taking advance of the specific JNK inhibitor peptide, D-JNKI1, we defined JNK action on the SNARE complex formation. Finally, electrophysiological recordings confirmed the role of JNK in the presynaptic modulation of vesicle release. These data suggest that JNK-dependent phosphorylation of T-SNARE proteins may have an important functional role in synaptic plasticity., Competing Interests: There is no actual or potential conflict of interests.
- Published
- 2017
- Full Text
- View/download PDF
11. Extended Synaptotagmin (ESyt) Triple Knock-Out Mice Are Viable and Fertile without Obvious Endoplasmic Reticulum Dysfunction.
- Author
-
Sclip A, Bacaj T, Giam LR, and Südhof TC
- Subjects
- Animals, Brain metabolism, Calcium metabolism, Calcium Signaling, Cell Line, Cell Survival genetics, Gene Knockout Techniques, Gene Order, Gene Targeting, Genetic Loci, Genotype, Humans, Mice, Mice, Knockout, Neurons metabolism, Stress, Physiological, Synaptotagmins genetics, Endoplasmic Reticulum metabolism, Fertility genetics, Phenotype, Synaptotagmins deficiency
- Abstract
Extended synaptotagmins (ESyts) are endoplasmic reticulum (ER) proteins composed of an N-terminal transmembrane region, a central SMP-domain, and five (ESyt1) or three C-terminal cytoplasmic C2-domains (ESyt2 and ESyt3). ESyts bind phospholipids in a Ca2+-dependent manner via their C2-domains, are localized to ER-plasma membrane contact sites, and may catalyze lipid exchange between the plasma membrane and the ER via their SMP-domains. However, the overall function of ESyts has remained enigmatic. Here, we generated triple constitutive and conditional knock-out mice that lack all three ESyt isoforms; in addition, we produced knock-in mice that express mutant ESyt1 or ESyt2 carrying inactivating substitutions in the Ca2+-binding sites of their C2A-domains. Strikingly, all ESyt mutant mice, even those lacking all ESyts, were apparently normal and survived and bred in a manner indistinguishable from control mice. ESyt mutant mice displayed no major changes in brain morphology or synaptic protein composition, and exhibited no large alterations in stress responses. Thus, in mice ESyts do not perform an essential role in basic cellular functions, suggesting that these highly conserved proteins may perform a specialized role that may manifest only during specific, as yet untested challenges.
- Published
- 2016
- Full Text
- View/download PDF
12. The cell-permeable Aβ1-6A2VTAT(D) peptide reverts synaptopathy induced by Aβ1-42wt.
- Author
-
Cimini S, Sclip A, Mancini S, Colombo L, Messa M, Cagnotto A, Di Fede G, Tagliavini F, Salmona M, and Borsello T
- Subjects
- Amyloid beta-Peptides ultrastructure, Animals, Cell Membrane Permeability, Dendritic Spines drug effects, Dendritic Spines metabolism, Disease Models, Animal, Hippocampus drug effects, Mice, Mice, Transgenic, Neurons drug effects, Peptide Fragments ultrastructure, Synapses drug effects, Alzheimer Disease metabolism, Amyloid beta-Peptides administration & dosage, Amyloid beta-Peptides toxicity, Hippocampus metabolism, Neurons metabolism, Peptide Fragments administration & dosage, Peptide Fragments toxicity, Synapses metabolism
- Abstract
Alzheimer disease (AD) is the most prevalent form of dementia. Loss of hippocampal synapses is the first neurodegenerative event in AD. Synaptic loss has been associated with the accumulation in the brain parenchyma of soluble oligomeric forms of amyloid β peptide (Aβ1-42wt). Clinical observations have shown that a mutation in the APP protein (A673V) causes an early onset AD-type dementia in homozygous carriers while heterozygous carriers are unaffected. This mutation leads to the formation of mutated Aβ peptides (Aβ1-42A2V) in homozygous patients, while in heterozygous subjects both Aβ1-42wt and Aβ1-42A2V are present. To better understand the impact of the A673V mutation in AD, we analyzed the synaptotoxic effect of oligomers formed by aggregation of different Aβ peptides (Aβ1-42wt or Aβ1-42A2V) and the combination of the two Aβ1-42MIX (Aβ1-42wt and Aβ1-42A2V) in an in vitro model of synaptic injury. We showed that Aβ1-42A2V oligomers are more toxic than Aβ1-42wt oligomers in hippocampal neurons, confirming the results previously obtained in cell lines. Furthermore, we reported that oligomers obtained by the combination of both wild type and mutated peptides (Aβ1-42MIX) did not exert synaptic toxicity. We concluded that the combination of Aβ1-42wt and Aβ1-42A2V peptides hinders the toxicity of Aβ1-42A2V and counteracts the manifestation of synaptopathy in vitro. Finally we took advantage of this finding to generate a cell-permeable peptide for clinical application, by fusing the first six residues of the Aβ1-42A2V to the TAT cargo sequence (Aβ1-6A2VTAT(D)). Noteworthy, the treatment with Aβ1-6A2VTAT(D) confers neuroprotection against both in vitro and in vivo synaptopathy models. Therefore Aβ1-6A2VTAT(D) may represent an innovative therapeutic tool to prevent synaptic degeneration in AD., (Copyright © 2015. Published by Elsevier Inc.)
- Published
- 2016
- Full Text
- View/download PDF
13. Exploring the role of MKK7 in excitotoxicity and cerebral ischemia: a novel pharmacological strategy against brain injury.
- Author
-
Vercelli A, Biggi S, Sclip A, Repetto IE, Cimini S, Falleroni F, Tomasi S, Monti R, Tonna N, Morelli F, Grande V, Stravalaci M, Biasini E, Marin O, Bianco F, di Marino D, and Borsello T
- Subjects
- Amino Acid Sequence, Animals, Animals, Newborn, Antigens, Differentiation chemistry, Antigens, Differentiation genetics, Antigens, Differentiation metabolism, Cell Hypoxia, Cerebral Cortex drug effects, Cerebral Cortex metabolism, Cerebral Cortex pathology, Electrocoagulation, Gene Expression Regulation, Glucose toxicity, Infarction, Middle Cerebral Artery genetics, Infarction, Middle Cerebral Artery metabolism, Infarction, Middle Cerebral Artery pathology, JNK Mitogen-Activated Protein Kinases genetics, JNK Mitogen-Activated Protein Kinases metabolism, MAP Kinase Kinase 7 chemistry, MAP Kinase Kinase 7 genetics, MAP Kinase Kinase 7 metabolism, Male, Molecular Docking Simulation, Molecular Sequence Data, N-Methylaspartate toxicity, Neurons metabolism, Neurons pathology, Neuroprotective Agents chemical synthesis, Peptides chemical synthesis, Primary Cell Culture, Protein Engineering, Rats, Rats, Sprague-Dawley, Signal Transduction, Thromboembolism, Tissue Culture Techniques, Infarction, Middle Cerebral Artery drug therapy, MAP Kinase Kinase 7 antagonists & inhibitors, Neurons drug effects, Neuroprotective Agents pharmacology, Peptides pharmacology
- Abstract
Excitotoxicity following cerebral ischemia elicits a molecular cascade, which leads to neuronal death. c-Jun-N-terminal kinase (JNK) has a key role in excitotoxic cell death. We have previously shown that JNK inhibition by a specific cell-permeable peptide significantly reduces infarct size and neuronal death in an in vivo model of cerebral ischemia. However, systemic inhibition of JNK may have detrimental side effects, owing to blockade of its physiological function. Here we designed a new inhibitor peptide (growth arrest and DNA damage-inducible 45β (GADD45β-I)) targeting mitogen-activated protein kinase kinase 7 (MKK7), an upstream activator of JNK, which exclusively mediates JNK's pathological activation. GADD45β-I was engineered by optimizing the domain of the GADD45β, able to bind to MKK7, and by linking it to the TAT peptide sequence, to allow penetration of biological membranes. Our data clearly indicate that GADD45β-I significantly reduces neuronal death in excitotoxicity induced by either N-methyl-D-aspartate exposure or by oxygen-glucose deprivation in vitro. Moreover, GADD45β-I exerted neuroprotection in vivo in two models of ischemia, obtained by electrocoagulation and by thromboembolic occlusion of the middle cerebral artery (MCAo). Indeed, GADD45β-I reduced the infarct size when injected 30 min before the lesion in both models. The peptide was also effective when administrated 6 h after lesion, as demonstrated in the electrocoagulation model. The neuroprotective effect of GADD45β-I is long lasting; in fact, 1 week after MCAo the infarct volume was still reduced by 49%. Targeting MKK7 could represent a new therapeutic strategy for the treatment of ischemia and other pathologies involving MKK7/JNK activation. Moreover, this new inhibitor can be useful to further dissect the physiological and pathological role of the JNK pathway in the brain.
- Published
- 2015
- Full Text
- View/download PDF
14. Region- and age-dependent reductions of hippocampal long-term potentiation and NMDA to AMPA ratio in a genetic model of Alzheimer's disease.
- Author
-
Tozzi A, Sclip A, Tantucci M, de Iure A, Ghiglieri V, Costa C, Di Filippo M, Borsello T, and Calabresi P
- Subjects
- Alzheimer Disease therapy, Amyloid beta-Protein Precursor genetics, Animals, Cholinesterase Inhibitors pharmacology, Disease Models, Animal, Long-Term Potentiation drug effects, Mice, Transgenic, Molecular Targeted Therapy, Mutation, Neostigmine pharmacology, Plaque, Amyloid metabolism, Aging genetics, Aging physiology, Alzheimer Disease genetics, Alzheimer Disease physiopathology, Hippocampus metabolism, Hippocampus physiopathology, Long-Term Potentiation genetics, N-Methylaspartate metabolism, alpha-Amino-3-hydroxy-5-methyl-4-isoxazolepropionic Acid metabolism
- Abstract
To characterize the mechanisms underlying region- and age-dependent hippocampal synaptic dysfunction in Alzheimer's disease, we used transgenic CRND8 mice, expressing the Swedish-Indiana APP mutation. In 2-month-old mice, no β-amyloid plaques deposition, but the presence of soluble oligomers, were found in CA1 area but not in dentate gyrus (DG). At this age, long-term potentiation (LTP) was reduced selectively in CA1. In 6-month-old mice, the presence of soluble oligomers was accompanied by accumulation of β-amyloid plaques and decreased LTP in CA1 and DG regions. In both regions, the loss of LTP was linked to reduced N-methyl-D-aspartate (NMDA) to alpha-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) current ratio. The acetylcholine-esterase inhibitor, neostigmine rescued LTP in CA1 area at early stage of the disease but not after plaques deposition. Conversely, the NMDA receptor antagonist memantine restored LTP selectively in DG at later stages of the disease. Both these effects were associated with a normalization of the NMDA to AMPA ratio. The association between the recovery of LTP and the normalization of the NMDA to AMPA ratio provides information on new possible therapeutic strategies in Alzheimer's disease., (Copyright © 2015 Elsevier Inc. All rights reserved.)
- Published
- 2015
- Full Text
- View/download PDF
15. Determination of tissue levels of a neuroprotectant drug: the cell permeable JNK inhibitor peptide.
- Author
-
Davoli E, Sclip A, Cecchi M, Cimini S, Carrà A, Salmona M, and Borsello T
- Subjects
- Animals, Animals, Newborn, Blood-Brain Barrier metabolism, Brain metabolism, Cell Membrane drug effects, Cell Membrane metabolism, Kidney metabolism, Liver metabolism, Mice, Neurons drug effects, Peptides antagonists & inhibitors, Cell Membrane Permeability drug effects, Neuroprotective Agents metabolism, Neuroprotective Agents pharmacology, Peptides metabolism, Peptides pharmacology
- Abstract
Introduction: Cell permeable peptides (CPPs) represent a novel tool for the delivery of bioactive molecules into scarcely accessible organs, such as the brain. CPPs have been successfully used in pre-clinical studies for a variety of diseases, ranging from cancer to neurological disorders. However, the mechanisms by which CPPs cross biological membranes, as well as their pharmacokinetic properties, have been poorly explored due to the lack of specific and sensitive analytical methods., Methods: In this paper we describe a protocol to quantitatively determine the amount of CPPs in in vitro and in vivo experimental models. To this end we selected the peptide D-JNKI1 that was shown to prevent neurodegeneration in both acute and chronic degenerative disorders. This method allows an accurate quantitative analysis of D-JNKI1 in both neuronal lysates and tissue homogenates using mass spectrometry and stable isotope dilution approach., Results: We found that D-JNKI1 crosses cellular membranes with fast kinetics, through an active and passive mechanism. After acute intraperitoneal (ip) administration of D-JNKI1 in mice, the peptide was found in the main organs with particular regard to the liver and kidney. Interestingly, D-JNKI1 crosses the blood brain barrier (BBB) and reaches the brain, where it remains for one week., Discussion: The challenge lies in developing the clinical application of therapeutic cell permeable peptides. Discerning pharmacokinetic properties is a high priority to produce a powerful therapeutic strategy. Overall, our data shed light on the pharmacokinetic properties of D-JNKI1 and supports its powerful neuroprotective effect., (Copyright © 2014 Elsevier Inc. All rights reserved.)
- Published
- 2014
- Full Text
- View/download PDF
16. c-Jun N-terminal kinase has a key role in Alzheimer disease synaptic dysfunction in vivo.
- Author
-
Sclip A, Tozzi A, Abaza A, Cardinetti D, Colombo I, Calabresi P, Salmona M, Welker E, and Borsello T
- Subjects
- Alzheimer Disease drug therapy, Alzheimer Disease genetics, Animals, Disease Models, Animal, Female, Humans, JNK Mitogen-Activated Protein Kinases antagonists & inhibitors, JNK Mitogen-Activated Protein Kinases genetics, Male, Mice, Mice, Transgenic, Peptides administration & dosage, Signal Transduction, Alzheimer Disease enzymology, JNK Mitogen-Activated Protein Kinases metabolism, Synapses enzymology
- Abstract
Altered synaptic function is considered one of the first features of Alzheimer disease (AD). Currently, no treatment is available to prevent the dysfunction of excitatory synapses in AD. Identification of the key modulators of synaptopathy is of particular significance in the treatment of AD. We here characterized the pathways leading to synaptopathy in TgCRND8 mice and showed that c-Jun N-terminal kinase (JNK) is activated at the spine prior to the onset of cognitive impairment. The specific inhibition of JNK, with its specific inhibiting peptide D-JNKI1, prevented synaptic dysfunction in TgCRND8 mice. D-JNKI1 avoided both the loss of postsynaptic proteins and glutamate receptors from the postsynaptic density and the reduction in size of excitatory synapses, reverting their dysfunction. This set of data reveals that JNK is a key signaling pathway in AD synaptic injury and that its specific inhibition offers an innovative therapeutic strategy to prevent spine degeneration in AD.
- Published
- 2014
- Full Text
- View/download PDF
17. Soluble Aβ oligomer-induced synaptopathy: c-Jun N-terminal kinase's role.
- Author
-
Sclip A, Arnaboldi A, Colombo I, Veglianese P, Colombo L, Messa M, Mancini S, Cimini S, Morelli F, Antoniou X, Welker E, Salmona M, and Borsello T
- Subjects
- Alzheimer Disease, Amyloid beta-Peptides pharmacology, Animals, Caspase 3 metabolism, Caspase 3 physiology, Dendritic Spines drug effects, Dendritic Spines ultrastructure, Humans, JNK Mitogen-Activated Protein Kinases metabolism, Mice, Models, Biological, Receptors, AMPA metabolism, Receptors, N-Methyl-D-Aspartate metabolism, Signal Transduction, Amyloid beta-Peptides metabolism, Dendritic Spines metabolism, JNK Mitogen-Activated Protein Kinases physiology
- Published
- 2013
- Full Text
- View/download PDF
18. An N-terminal fragment of the prion protein binds to amyloid-β oligomers and inhibits their neurotoxicity in vivo.
- Author
-
Fluharty BR, Biasini E, Stravalaci M, Sclip A, Diomede L, Balducci C, La Vitola P, Messa M, Colombo L, Forloni G, Borsello T, Gobbi M, and Harris DA
- Subjects
- Alzheimer Disease metabolism, Amyloidogenic Proteins chemistry, Animals, Benzothiazoles, Caenorhabditis elegans metabolism, Hippocampus metabolism, Mice, Mice, Inbred C57BL, Models, Biological, Neurodegenerative Diseases metabolism, Neurons metabolism, Peptides chemistry, Protein Binding, Protein Structure, Tertiary, Surface Plasmon Resonance, Synapses metabolism, Thiazoles chemistry, Amyloid beta-Peptides chemistry, Prions chemistry
- Abstract
A hallmark of Alzheimer disease (AD) is the accumulation of the amyloid-β (Aβ) peptide in the brain. Considerable evidence suggests that soluble Aβ oligomers are responsible for the synaptic dysfunction and cognitive deficit observed in AD. However, the mechanism by which these oligomers exert their neurotoxic effect remains unknown. Recently, it was reported that Aβ oligomers bind to the cellular prion protein with high affinity. Here, we show that N1, the main physiological cleavage fragment of the cellular prion protein, is necessary and sufficient for binding early oligomeric intermediates during Aβ polymerization into amyloid fibrils. The ability of N1 to bind Aβ oligomers is influenced by positively charged residues in two sites (positions 23-31 and 95-105) and is dependent on the length of the sequence between them. Importantly, we also show that N1 strongly suppresses Aβ oligomer toxicity in cultured murine hippocampal neurons, in a Caenorhabditis elegans-based assay, and in vivo in a mouse model of Aβ-induced memory dysfunction. These data suggest that N1, or small peptides derived from it, could be potent inhibitors of Aβ oligomer toxicity and represent an entirely new class of therapeutic agents for AD.
- Published
- 2013
- Full Text
- View/download PDF
19. The neurodegeneration in Alzheimer disease and the prion protein.
- Author
-
Forloni G, Sclip A, Borsello T, and Balducci C
- Subjects
- Amyloid beta-Peptides analysis, Amyloid beta-Peptides toxicity, Animals, Brain metabolism, Humans, Prions analysis, Alzheimer Disease metabolism, Alzheimer Disease pathology, Amyloid beta-Peptides metabolism, Brain pathology, Prions metabolism
- Abstract
The concept of "prion-like" has been proposed to explain the pathogenic mechanism of the principal neurodegenerative disorders associated with protein misfolding, including Alzheimer disease (AD). Other evidence relates prion protein with AD: the cellular prion protein (PrP(C)) binds β amyloid oligomers, allegedly responsible for the neurodegeneration in AD, mediating their toxic effects. We and others have confirmed the high-affinity binding between β amyloid oligomers and PrP(C), but we were not able to assess the functional consequences of this interaction using behavioral investigations and in vitro tests. This discrepancy rather than being resolved with the classic explanations, differencies in methodological aspects, has been reinforced by new data from different sources. Here we present data obtained with PrP antibody that not interfere with the neurotoxic activity of β amyloid oligomers. Since the potential role of the PrP(C) in the neuronal dysfunction induced by β amyloid oligomers is an important issue, find reasonable explanation of the inconsistent results is needed. Even more important however is the relevance of this interaction in the context of the disease, so as to develop valid therapeutic strategies.
- Published
- 2013
- Full Text
- View/download PDF
20. Specific inhibition of the JNK pathway promotes locomotor recovery and neuroprotection after mouse spinal cord injury.
- Author
-
Repici M, Chen X, Morel MP, Doulazmi M, Sclip A, Cannaya V, Veglianese P, Kraftsik R, Mariani J, Borsello T, and Dusart I
- Subjects
- Animals, Blood Vessels drug effects, Blood Vessels physiology, Blotting, Western, Caspase 3 metabolism, Hindlimb physiology, Image Processing, Computer-Assisted, Imaging, Three-Dimensional, Immunohistochemistry, Injections, Intraperitoneal, Male, Mice, Nerve Fibers physiology, Protein Kinase Inhibitors administration & dosage, Proto-Oncogene Proteins c-jun metabolism, Serotonin physiology, Spinal Cord pathology, Spinal Cord Injuries enzymology, Spinal Cord Injuries physiopathology, JNK Mitogen-Activated Protein Kinases antagonists & inhibitors, Locomotion drug effects, Neuroprotective Agents, Peptides pharmacology, Protein Kinase Inhibitors pharmacology, Recovery of Function drug effects, Signal Transduction drug effects, Spinal Cord Injuries drug therapy
- Abstract
Limiting the development of secondary damage represents one of the major goals of neuroprotective therapies after spinal cord injury. Here, we demonstrate that specific JNK inhibition via a single intraperitoneal injection of the cell permeable peptide D-JNKI1 6h after lesion improves locomotor recovery assessed by both the footprint and the BMS tests up to 4 months post-injury in mice. JNK inhibition prevents c-jun phosphorylation and caspase-3 cleavage, has neuroprotective effects and results in an increased sparing of white matter at the lesion site. Lastly, D-JNKI1 treated animals show a lower increase of erythrocyte extravasation and blood brain barrier permeability, thus indicating protection of the vascular system. In total, these results clearly point out JNK inhibition as a promising neuroprotective strategy for preventing the evolution of secondary damage after spinal cord injury., (Copyright © 2012 Elsevier Inc. All rights reserved.)
- Published
- 2012
- Full Text
- View/download PDF
21. c-Jun N-terminal kinase regulates soluble Aβ oligomers and cognitive impairment in AD mouse model.
- Author
-
Sclip A, Antoniou X, Colombo A, Camici GG, Pozzi L, Cardinetti D, Feligioni M, Veglianese P, Bahlmann FH, Cervo L, Balducci C, Costa C, Tozzi A, Calabresi P, Forloni G, and Borsello T
- Subjects
- Alzheimer Disease genetics, Animals, Cognition Disorders metabolism, Disease Models, Animal, Electrophysiology, Humans, Maze Learning, Memory Disorders genetics, Mice, Models, Biological, Peptides chemistry, Signal Transduction, Time Factors, Alzheimer Disease metabolism, Amyloid beta-Peptides chemistry, Gene Expression Regulation, Enzymologic, JNK Mitogen-Activated Protein Kinases metabolism
- Abstract
Alzheimer disease (AD) is characterized by cognitive impairment that starts with memory loss to end in dementia. Loss of synapses and synaptic dysfunction are closely associated with cognitive impairment in AD patients. Biochemical and pathological evidence suggests that soluble Aβ oligomers correlate with cognitive impairment. Here, we used the TgCRND8 AD mouse model to investigate the role of JNK in long term memory deficits. TgCRND8 mice were chronically treated with the cell-penetrating c-Jun N-terminal kinase inhibitor peptide (D-JNKI1). D-JNKI1, preventing JNK action, completely rescued memory impairments (behavioral studies) as well as the long term potentiation deficits of TgCRND8 mice. Moreover, D-JNKI1 inhibited APP phosphorylation in Thr-668 and reduced the amyloidogenic cleavage of APP and Aβ oligomers in brain parenchyma of treated mice. In conclusion, by regulating key pathogenic mechanisms of AD, JNK might hold promise as innovative therapeutic target.
- Published
- 2011
- Full Text
- View/download PDF
22. JNK plays a key role in tau hyperphosphorylation in Alzheimer's disease models.
- Author
-
Ploia C, Antoniou X, Sclip A, Grande V, Cardinetti D, Colombo A, Canu N, Benussi L, Ghidoni R, Forloni G, and Borsello T
- Subjects
- Aged, Aged, 80 and over, Alzheimer Disease pathology, Animals, Cells, Cultured, Cerebral Cortex drug effects, Cerebral Cortex pathology, Female, Humans, JNK Mitogen-Activated Protein Kinases pharmacology, Male, Middle Aged, Mitogen-Activated Protein Kinases metabolism, Neurofibrillary Tangles drug effects, Neurofibrillary Tangles metabolism, Neurofibrillary Tangles pathology, Neurons drug effects, Neurons pathology, Phosphorylation, Rats, Alzheimer Disease metabolism, Cerebral Cortex metabolism, JNK Mitogen-Activated Protein Kinases metabolism, Neurons metabolism, tau Proteins metabolism
- Abstract
Alzheimer's disease (AD) is a major clinical concern, and the search for new molecules to combat disease progression remains important. One of the major hallmarks in AD pathogenesis is the hyperphosphorylation of tau and subsequent formation of neurofibrillary tangles. Several kinases are involved in this process. Amongst them, c-Jun N-terminal kinases (JNKs) are activated in AD brains and are also associated with the development of amyloid plaques. This study was designed to investigate the contribution of JNK in tau hyperphosphorylation and whether it may represent a potential therapeutic target for the fight against AD. The specific inhibition of JNK by the cell permeable peptide D-JNKI-1 led to a reduction of p-tau at S202/T205 and S422, two established target sites of JNK, in rat neuronal cultures and in human fibroblasts cultures. Similarly, D-JNKI-1 reduced p-tau at S202/T205 in an in vivo model of AD (TgCRND8 mice). Our findings support the fundamental role of JNK in the regulation of tau hyperphosphorylation and subsequently in AD pathogenesis.
- Published
- 2011
- Full Text
- View/download PDF
23. Crosstalk between JNK and SUMO signaling pathways: deSUMOylation is protective against H2O2-induced cell injury.
- Author
-
Feligioni M, Brambilla E, Camassa A, Sclip A, Arnaboldi A, Morelli F, Antoniou X, and Borsello T
- Subjects
- Cell Death, Cell Line, Tumor, Cell Survival, Enzyme Activation, Gene Expression Regulation, Enzymologic, Humans, Immunohistochemistry methods, Immunoprecipitation, Oxidative Stress, Plasmids metabolism, Proto-Oncogene Proteins c-jun metabolism, Signal Transduction, Tetrazolium Salts pharmacology, Thiazoles pharmacology, Time Factors, Hydrogen Peroxide metabolism, JNK Mitogen-Activated Protein Kinases metabolism, Small Ubiquitin-Related Modifier Proteins metabolism
- Abstract
Background: Oxidative stress is a key feature in the pathogenesis of several neurological disorders. Following oxidative stress stimuli a wide range of pathways are activated and contribute to cellular death. The mechanism that couples c-Jun N-terminal kinase (JNK) signaling, a key pathway in stress conditions, to the small ubiquitin-related modifier (SUMO), an emerging protein in the field, is largely unknown., Methodology/principal Findings: With this study we investigated if SUMOylation participates in the regulation of JNK activation as well as cellular death in a model of H(2)O(2) induced-oxidative stress. Our data show that H(2)O(2) modulates JNK activation and induces cellular death in neuroblastoma SH-SY5Y cells. Inhibition of JNK's action with the D-JNKI1 peptide rescued cells from death. Following H(2)O(2), SUMO-1 over-expression increased phosphorylation of JNK and exacerbated cell death, although only in conditions of mild oxidative stress. Furthermore inhibition of SUMOylation, following transfection with SENP1, interfered with JNK activation and rescued cells from H(2)O(2) induced death. Importantly, in our model, direct interaction between these proteins can occur., Conclusions/significance: Taken together our results show that SUMOylation may significantly contribute to modulation of JNK activation and contribute to cell death in oxidative stress conditions.
- Published
- 2011
- Full Text
- View/download PDF
24. Synthetic amyloid-beta oligomers impair long-term memory independently of cellular prion protein.
- Author
-
Balducci C, Beeg M, Stravalaci M, Bastone A, Sclip A, Biasini E, Tapella L, Colombo L, Manzoni C, Borsello T, Chiesa R, Gobbi M, Salmona M, and Forloni G
- Subjects
- Alzheimer Disease etiology, Amyloid beta-Peptides chemical synthesis, Amyloid beta-Peptides chemistry, Animals, Cognition Disorders etiology, Cognition Disorders metabolism, Humans, Injections, Intraventricular, Male, Memory physiology, Mice, Mice, Inbred C57BL, Mice, Knockout, Neuronal Plasticity drug effects, Neuronal Plasticity physiology, Peptide Fragments chemical synthesis, Peptide Fragments chemistry, Prion Proteins, Prions genetics, Prions metabolism, Protein Binding, Surface Plasmon Resonance, Amyloid beta-Peptides pharmacology, Memory drug effects, Peptide Fragments pharmacology, PrPC Proteins metabolism
- Abstract
Inability to form new memories is an early clinical sign of Alzheimer's disease (AD). There is ample evidence that the amyloid-beta (Abeta) peptide plays a key role in the pathogenesis of this disorder. Soluble, bio-derived oligomers of Abeta are proposed as the key mediators of synaptic and cognitive dysfunction, but more tractable models of Abeta-mediated cognitive impairment are needed. Here we report that, in mice, acute intracerebroventricular injections of synthetic Abeta(1-42) oligomers impaired consolidation of the long-term recognition memory, whereas mature Abeta(1-42) fibrils and freshly dissolved peptide did not. The deficit induced by oligomers was reversible and was prevented by an anti-Abeta antibody. It has been suggested that the cellular prion protein (PrP(C)) mediates the impairment of synaptic plasticity induced by Abeta. We confirmed that Abeta(1-42) oligomers interact with PrP(C), with nanomolar affinity. However, PrP-expressing and PrP knock-out mice were equally susceptible to this impairment. These data suggest that Abeta(1-42) oligomers are responsible for cognitive impairment in AD and that PrP(C) is not required.
- Published
- 2010
- Full Text
- View/download PDF
25. Role of Glycogen Synthase Kinase-3β in APP Hyperphosphorylation Induced by NMDA Stimulation in Cortical Neurons.
- Author
-
Ploia C, Sclip A, Colombo A, Repici M, Gardoni F, Di Luca M, Forloni G, Antoniou X, and Borsello T
- Abstract
The phosphorylation of Amyloid Precursor Protein (APP) at Thr
668 plays a key role in APP metabolism that is highly relevant to AD. The c-Jun-N-terminal kinase (JNK), glycogen synthase kinase-3β (GSK-3β) and cyclin-dependent kinase 5 (Cdk5) can all be responsible for this phosphorylation. These kinases are activated by excitotoxic stimuli fundamental hallmarks of AD. The exposure of cortical neurons to a high dose of NMDA (100 μM) for 30'-45' led to an increase of P-APP Thr668 . During NMDA stimulation APP hyperphosphorylation has to be assigned to GSK-3β activity, since addition of L803-mts, a substrate competitive inhibitor of GSK-3β reduced APP phosphorylation induced by NMDA. On the contrary, inhibition of JNK and Cdk5 with D-JNKI1 and Roscovitine respectively did not prevent NMDA-induced P-APP increase. These data show a tight connection, in excitotoxic conditions, between APP metabolism and the GSK-3β signaling pathway.- Published
- 2010
- Full Text
- View/download PDF
26. JNK contributes to Hif-1alpha regulation in hypoxic neurons.
- Author
-
Antoniou X, Sclip A, Ploia C, Colombo A, Moroy G, and Borsello T
- Subjects
- Aging pathology, Amino Acid Sequence, Animals, Cell Death, Cell Hypoxia, Humans, Hypoxia-Inducible Factor 1, alpha Subunit chemistry, MAP Kinase Signaling System, Molecular Sequence Data, Protein Binding, Protein Structure, Secondary, Protein Structure, Tertiary, Rats, Hypoxia-Inducible Factor 1, alpha Subunit metabolism, JNK Mitogen-Activated Protein Kinases metabolism, Neurons enzymology, Neurons pathology
- Abstract
Hypoxia is an established factor of neurodegeneration. Nowadays, attention is directed at understanding how alterations in the expression of stress-related signaling proteins contribute to age dependent neuronal vulnerability to injury. The purpose of this study was to investigate how Hif-1alpha, a major neuroprotective factor, and JNK signaling, a key pathway in neurodegeneration, relate to hypoxic injury in young (6DIV) and adult (12DIV) neurons. We could show that in young neurons as compared to mature ones, the protective factor Hif-1alpha is more induced while the stress protein phospho-JNK displays lower basal levels. Indeed, changes in the expression levels of these proteins correlated with increased vulnerability of adult neurons to hypoxic injury. Furthermore, we describe for the first time that treatment with the D-JNKI1, a JNK-inhibiting peptide, rescues adult hypoxic neurons from death and contributes to Hif-1alpha upregulation, probably via a direct interaction with the Hif-1alpha protein.
- Published
- 2009
- Full Text
- View/download PDF
27. c-Jun N-terminal kinase pathway activation in human and experimental cerebral contusion.
- Author
-
Ortolano F, Colombo A, Zanier ER, Sclip A, Longhi L, Perego C, Stocchetti N, Borsello T, and De Simoni MG
- Subjects
- Adult, Aged, Animals, Blotting, Western, Brain Injuries pathology, Enzyme Activation drug effects, Enzyme Inhibitors pharmacology, Female, Humans, In Situ Nick-End Labeling, JNK Mitogen-Activated Protein Kinases drug effects, Male, Mice, Mice, Inbred C57BL, Middle Aged, Signal Transduction drug effects, Tomography, X-Ray Computed, Brain Injuries enzymology, Enzyme Activation physiology, JNK Mitogen-Activated Protein Kinases metabolism, Signal Transduction physiology
- Abstract
The c-Jun N-terminal kinase (JNK) pathway is involved in cell stress and apoptosis. We tested the hypothesis that this pathway plays a role in traumatic brain injury (TBI) by assessing JNK activation in human brain tissues and in brains of mice subjected to controlled cortical impact brain injury. We also assessed the effects of specific inhibition of the JNK pathway by the cell-permeable JNK inhibitor peptide, D-JNKI1, on neurobehavioral function and posttraumatic cell loss in mice. The inhibitor was administered intraperitoneally 10 minutes after injury. The JNK pathway showed robust activation both in human contusion specimens and in injured cortex and hippocampi of TBI-injured mice, 1, 4, and 48 hours after injury. D-JNKI1 treatment significantly improved motor performance at 48 hours and 7 days after injury and reduced the contusion volume compared with saline treatment; the numbers of terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling-positive cells were significantly decreased in the hippocampi of injured mice 48 hours after treatment. Thus, because the JNK pathway is activated after human and experimental TBI and the inhibitor peptide D-JNKI1 affords significant neuroprotection and amelioration of neurobehavioral deficits after experimental TBI, therapeutic targeting of the JNK activation pathway may hold promise for future clinical applications.
- Published
- 2009
- Full Text
- View/download PDF
28. c-Jun N-terminal kinase binding domain-dependent phosphorylation of mitogen-activated protein kinase kinase 4 and mitogen-activated protein kinase kinase 7 and balancing cross-talk between c-Jun N-terminal kinase and extracellular signal-regulated kinase pathways in cortical neurons.
- Author
-
Repici M, Mare L, Colombo A, Ploia C, Sclip A, Bonny C, Nicod P, Salmona M, and Borsello T
- Subjects
- Activating Transcription Factor 2 metabolism, Amino Acid Sequence, Analysis of Variance, Animals, Animals, Newborn, Dose-Response Relationship, Drug, Enzyme Activation drug effects, Extracellular Signal-Regulated MAP Kinases, L-Lactate Dehydrogenase metabolism, Peptides pharmacology, Phosphorylation, Protein Binding physiology, Protein Interaction Domains and Motifs, Rats, Signal Transduction drug effects, ets-Domain Protein Elk-1 metabolism, Cerebral Cortex cytology, JNK Mitogen-Activated Protein Kinases metabolism, MAP Kinase Kinase 4 metabolism, MAP Kinase Kinase 7 metabolism, Neurons metabolism, Signal Transduction physiology
- Abstract
The c-Jun N-terminal kinase (JNK) is a mitogen-activated protein kinase (MAPK) activated by stress-signals and involved in many different diseases. Previous results proved the powerful effect of the cell permeable peptide inhibitor d-JNKI1 (d-retro-inverso form of c-Jun N-terminal kinase-inhibitor) against neuronal death in CNS diseases, but the precise features of this neuroprotection remain unclear. We here performed cell-free and in vitro experiments for a deeper characterization of d-JNKI1 features in physiological conditions. This peptide works by preventing JNK interaction with its c-Jun N-terminal kinase-binding domain (JBD) dependent targets. We here focused on the two JNK upstream MAPKKs, mitogen-activated protein kinase kinase 4 (MKK4) and mitogen-activated protein kinase kinase 7 (MKK7), because they contain a JBD homology domain. We proved that d-JNKI1 prevents MKK4 and MKK7 activity in cell-free and in vitro experiments: these MAPKK could be considered not only activators but also substrates of JNK. This means that d-JNKI1 can interrupt downstream but also upstream events along the JNK cascade, highlighting a new remarkable feature of this peptide. We also showed the lack of any direct effect of the peptide on p38, MEK1, and extracellular signal-regulated kinase (ERK) in cell free, while in rat primary cortical neurons JNK inhibition activates the MEK1-ERK-Ets1/c-Fos cascade. JNK inhibition induces a compensatory effect and leads to ERK activation via MEK1, resulting in an activation of the survival pathway-(MEK1/ERK) as a consequence of the death pathway-(JNK) inhibition. This study should hold as an important step to clarify the strong neuroprotective effect of d-JNKI1.
- Published
- 2009
- Full Text
- View/download PDF
29. JNK regulates APP cleavage and degradation in a model of Alzheimer's disease.
- Author
-
Colombo A, Bastone A, Ploia C, Sclip A, Salmona M, Forloni G, and Borsello T
- Subjects
- Alzheimer Disease genetics, Amyloid Precursor Protein Secretases metabolism, Amyloid beta-Protein Precursor genetics, Aspartic Acid Endopeptidases metabolism, Cell Line, Tumor, Cell Survival, Enzyme-Linked Immunosorbent Assay, Fluoroimmunoassay, Humans, Immunoblotting, Immunohistochemistry, JNK Mitogen-Activated Protein Kinases antagonists & inhibitors, MAP Kinase Signaling System, Mutation, Peptide Fragments metabolism, Peptides metabolism, Peptides pharmacology, Phosphorylation, Protease Nexins, Receptors, Cell Surface genetics, Alzheimer Disease metabolism, Amyloid beta-Protein Precursor metabolism, JNK Mitogen-Activated Protein Kinases metabolism, Receptors, Cell Surface metabolism
- Abstract
Secretion of Amyloid-beta peptide (Abeta) circulating oligomers and their aggregate forms derived by processing of beta-amyloid precursor protein (APP) are a key event in Alzheimer's disease (AD). We show that phosphorylation of APP on threonine 668 may play a role in APP metabolism in H4-APP(sw) cell line, a degenerative AD model. We proved that JNK plays a fundamental role in this phosphorylation since its specific inhibition, with the JNK inhibitor peptide (D-JNKI1), induced APP degradation and prevented APP phosphorylation at T668. This results in a significant drop of betaAPPs, Abeta fragments and Abeta circulating oligomers. Moreover the D-JNKI1 treatment produced a switch in the APP metabolism, since the peptide reduced the rate of the amyloidogenic processing in favour of the non-amyloidogenic one. All together our results suggest an important link between APP metabolism and the JNK pathway and contribute to shed light on the molecular signalling pathway of this disease indicating JNK as an innovative target for AD therapy.
- Published
- 2009
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.