132 results on '"Borsello T"'
Search Results
2. Principi di anatomia microscopica con cenni di istologia e anatomia macroscopica
- Author
-
Bertelli, E, Borsello, T, Campanella, C, Conconi, Mt, Di Liddo, R, Rumio, C, and Sommariva, M
- Subjects
Microscopia ,Anatomia ,Anatomia, Microscopia, Istologia ,Istologia - Published
- 2018
3. CEREBELLAR GRANULE CELLS AS A MODEL FOR ALZHEIMER DISEASE. AN IN VITRO STUDY ON ′-AMYLOID METABOLISM AND SECRETION IN APOPTOTIC NEURONS.
- Author
-
Galli, C., Piccini, A., Borsello, T., Ciotti, M T., Zaccheo, D., Tabaton, M., and Calissano, P.
- Published
- 1998
4. MOSSY FIBERS AXOTOMY INDUCES MASSIVE CEREBELLAR GRANULE NEURONS APOPTOSIS IN ADULT RATS: A LESIONAL MODEL FOR DEAFFERENTATION RELATED CELL DEATH.
- Author
-
Borsello, T., Ciotti, M T, Di Luzio, A., Calissano, P., and Galli, C.
- Published
- 1998
5. Targeting ß-amyloid by the A2V Aß variant: a novel disease-modifying strategy for the treatment of Alzheimer’s disease
- Author
-
Di Fede, G, Diomede, L, Catania, M, Maderna, E, Moda, F, Ruggerone, M, Romeo, M, Morbin, M, Palamara, L, Campagnani, I, Colombo, L, Rossi, A, Cagnotto, A, Messa, M, De Luigi, A, MANCINI, SIMONA, Stravalaci, M, Gobbi, M, Borsello, T, Salmona, M, Tagliavini, F., Di Fede, G, Diomede, L, Catania, M, Maderna, E, Moda, F, Ruggerone, M, Romeo, M, Morbin, M, Palamara, L, Campagnani, I, Colombo, L, Rossi, A, Cagnotto, A, Messa, M, De Luigi, A, Mancini, S, Stravalaci, M, Gobbi, M, Borsello, T, Salmona, M, and Tagliavini, F
- Subjects
Neurology ,Animal ,Medicine (all) ,Dementia ,Human - Published
- 2014
6. Contribution of serine racemase/d-serine pathway to neuronal apoptosis
- Author
-
Esposito, S, Pristerà, A, Maresca, G, Cavallaro, S, Felsani, A, Florenzano, F, Manni, L, Ciotti, M, Pollegioni, L, Borsello, T, and Canu, N
- Subjects
Proteasome Endopeptidase Complex ,Racemases and Epimerases ,Apoptosis ,serine.racemase ,In Vitro Techniques ,Settore BIO/09 ,Receptors, N-Methyl-D-Aspartate ,neuronal death, proteasome, c.Jun N-terminal kinase, NMDA receptor, serine.racemase, cerebellar granule neurons, apoptosis ,Cerebellum ,Serine ,Animals ,RNA, Messenger ,cerebellar granule neurons ,RNA, Small Interfering ,Rats, Wistar ,Cellular Senescence ,Neurons ,JNK Mitogen-Activated Protein Kinases ,Stereoisomerism ,NMDA receptor ,neuronal death ,Rats ,proteasome ,nervous system ,c.Jun N-terminal kinase ,Signal Transduction - Abstract
Recent data indicate that age-related N-methyl-d-aspartate receptor (NMDAR) transmission impairment is correlated with the reduction in serine racemase (SR) expression and d-serine content. As apoptosis is associated with several diseases and conditions that generally occur with age, we investigated the modulation of SR/d-serine pathway during neuronal apoptosis and its impact on survival. We found that in cerebellar granule neurons (CGNs), undergoing apoptosis SR/d-serine pathway is crucially regulated. In the early phase of apoptosis, the expression of SR is reduced, both at the protein and RNA level through pathways, upstream of caspase activation, involving ubiquitin proteasome system (UPS) and c-Jun N-terminal kinases (JNKs). Forced expression of SR, together with treatment with NMDA and d-serine, blocks neuronal death, whereas pharmacological inhibition and Sh-RNA-mediated suppression of endogenous SR exacerbate neuronal death. In the late phase of apoptosis, the increased expression of SR contribute to the last, NMDAR-mediated, wave of cell death. These findings are relevant to our understanding of neuronal apoptosis and NMDAR activity regulation, raising further questions as to the role of SR/d-serine in those neuro-pathophysiological processes, such as aging and neurodegenerative diseases characterized by a convergence of apoptotic mechanisms and NMDAR dysfunction.
- Published
- 2012
- Full Text
- View/download PDF
7. c-Jun N-terminal Kinase regulates soluble Abeta oligomers and cognitive impairment in an 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, Cinzia, Tozzi, Alessandro, Calabresi, Paolo, Forloni, G, and Borsello, T.
- Published
- 2011
8. Exploring the role of MKK7 in excitotoxicity and cerebral ischemia: a novel pharmacological strategy against brain injury
- Author
-
Vercelli, A, primary, Biggi, S, additional, Sclip, A, additional, Repetto, I E, additional, Cimini, S, additional, Falleroni, F, additional, Tomasi, S, additional, Monti, R, additional, Tonna, N, additional, Morelli, F, additional, Grande, V, additional, Stravalaci, M, additional, Biasini, E, additional, Marin, O, additional, Bianco, F, additional, di Marino, D, additional, and Borsello, T, additional
- Published
- 2015
- Full Text
- View/download PDF
9. Neuronal death and neuroprotection: a review
- Author
-
Repici, M., Jean Mariani, Borsello, T., Tchang, Francoise, Neurobiologie des processus adaptatifs ( NPA ), Université Pierre et Marie Curie - Paris 6 ( UPMC ) -Centre National de la Recherche Scientifique ( CNRS ), Département de Biologie Cellulaire et de Morphologie, Université de Lausanne ( UNIL ), Hôpital Charles Foix - Jean Rostand, Assistance publique - Hôpitaux de Paris (AP-HP)-Hôpital Charles Foix - Jean Rostand, Service d'explorations fonctionnelles, GIS, Institut de la longevite et du vieillissement, Neurobiologie des processus adaptatifs (NPA), Université Pierre et Marie Curie - Paris 6 (UPMC)-Centre National de la Recherche Scientifique (CNRS), Université de Lausanne (UNIL), CHU Charles Foix [AP-HP], Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Sorbonne Université (SU), Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Sorbonne Université (SU)-Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Sorbonne Université (SU), and Université de Lausanne = University of Lausanne (UNIL)
- Subjects
Brain Diseases ,Neuroprotective Agents ,nervous system ,Cell Death ,[SCCO.NEUR]Cognitive science/Neuroscience ,[ SCCO.NEUR ] Cognitive science/Neuroscience ,[SCCO.NEUR] Cognitive science/Neuroscience ,Autophagy ,Animals ,Humans ,Apoptosis - Abstract
To achieve neuroprotection is one of the main interests for neuroscientist: understanding the control mechanisms of neuronal death allows developing new tools for preventing it. Neuronal death plays a critical role in most of the important neural pathologies, including stroke, epilepsy, Parkinson's disease and Alzheimer's disease. This review summarizes the three main different types of neuronal death: apoptosis, necrosis and autophagic cell death, although we are conscious that if cell death falls into several categories, the boundaries are not always distinct. We then introduce the current understanding of the relationship between neuronal death types and neuroprotection.
- Published
- 2008
10. c-Jun N-terminal kinase has a key role in Alzheimer disease synaptic dysfunction in vivo
- Author
-
Sclip, A, primary, Tozzi, A, additional, Abaza, A, additional, Cardinetti, D, additional, Colombo, I, additional, Calabresi, P, additional, Salmona, M, additional, Welker, E, additional, and Borsello, T, additional
- Published
- 2014
- Full Text
- View/download PDF
11. Granule neuron DNA damage following differentation in adult rats cerebellar cortex: a lesional model
- Author
-
Borsello T., Di Luzio A., Ciotti M.T., Calissano P., and Galli C.
- Published
- 2000
12. The stem cells as a potential treatment for neurodegeneration
- Author
-
Borsello, T, Ferrari, D, Vescovi, A, Bottai, D, Daniela, Ferrari, Vescovi, Angelo Luigi, Bottai, Daniele, Borsello, T, Ferrari, D, Vescovi, A, Bottai, D, Daniela, Ferrari, Vescovi, Angelo Luigi, and Bottai, Daniele
- Abstract
Cell degeneration and death, be it extensive and widespread, such as in metabolic disorders, or focal and selective as in Parkinson's disease (PD), is the underlying feature of many neurological diseases. Thus, the replacement of cells lost by injury or disease has become a central tenet in strategies aiming at the development of novel therapeutic approaches for neurodegenerative disorders. In addition to the in vivo recruitment of endogenous cells, which is now emerging as a promising novel strategy, the transplantation of new, exogenously generated brain cells is probably the most extensively studied methodology for cell replacement in the central nervous system, with the initial experimental clinical studies in PD dating back to the early 1970s (Bjorklund, A. and Stenevi, U., 1984, Intracerebral neural implants: neuronal replacement and reconstruction of damaged circuitries. Annu Rev Neurosci 7, 279-308; Snyder, B. J. and Olanow, C. W., 2005, Stem cell treatment for Parkinson's disease: an update for 2005. Curr Opin Neurol 18, 376-85). The need to generate the cells to be transplanted in large quantities and in a reproducible, steady, and safe fashion has long represented one of the major issues in this field, regardless of whether one was trying to produce specific cell subtypes or uncommitted and highly plastic neural precursors, which would respond to local, instructive cues, upon transplantation into the damaged area. Neural stem cells (NSCs), with their capacity for long-term expansion in vitro and their extensive functional stability and plasticity, allow now for the establishment of cultures of mature neural cells as well as highly undifferentiated precursors and are emerging as one of the most amenable cell sources for neural transplantation (Gage, F. H., 2000, Mammalian neural stem cells. Science 287, 1433-8; McKay, R., 1997, Stem cells in the central nervous system. Science 276, 66-71). This chapter illustrates the basic aspect of the handling and prepa
- Published
- 2007
13. 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., primary, Mare, L., additional, Colombo, A., additional, Ploia, C., additional, Sclip, A., additional, Bonny, C., additional, Nicod, P., additional, Salmona, M., additional, and Borsello, T., additional
- Published
- 2009
- Full Text
- View/download PDF
14. c-Jun N-terminal kinase pathway activation in human and experimental traumatic brain injury: neuroprotective effects of its inhibition
- Author
-
Ortolano, F, primary, Zanier, ER, additional, Colombo, A, additional, Sclip, A, additional, Longhi, L, additional, Perego, C, additional, Borsello, T, additional, Stocchetti, N, additional, and De Simoni, MG, additional
- Published
- 2008
- Full Text
- View/download PDF
15. Time-course of c-Jun N-terminal kinase activation after cerebral ischemia and effect of D-JNKI1 on c-Jun and caspase-3 activation
- Author
-
Repici, M., primary, Centeno, C., additional, Tomasi, S., additional, Forloni, G., additional, Bonny, C., additional, Vercelli, A., additional, and Borsello, T., additional
- Published
- 2007
- Full Text
- View/download PDF
16. Phosphorylation‐dependent dimerization and subcellular localization of islet‐brain 1/c‐Jun N‐terminal kinase‐interacting protein 1
- Author
-
Borsello, T., primary, Centeno, C., additional, Riederer, I.M., additional, Haefliger, J.‐A., additional, and Riederer, B.M., additional
- Published
- 2007
- Full Text
- View/download PDF
17. The TAT-JNK inhibitor peptide interferes with beta amyloid protein stability
- Author
-
Colombo, A, primary, Repici, M, additional, Pesaresi, M, additional, Santambrogio, S, additional, Forloni, G, additional, and Borsello, T, additional
- Published
- 2007
- Full Text
- View/download PDF
18. C-JUN NH2-TERMINAL KINASE MEDIATES THE HEPATIC STRESS RESPONSE AFTER RAT HEMORRHAGE/RESUSCITATION
- Author
-
Relja, B., primary, Schwestka, B., additional, Sun-Young Lee, V., additional, Henrich, D., additional, Czerny, C., additional, Borsello, T., additional, Lehnert, M., additional, and Marzi, I., additional
- Published
- 2006
- Full Text
- View/download PDF
19. Role of the JNK pathway in NMDA-mediated excitotoxicity of cortical neurons
- Author
-
Centeno, C, primary, Repici, M, additional, Chatton, J-Y, additional, Riederer, B M, additional, Bonny, C, additional, Nicod, P, additional, Price, M, additional, Clarke, P G H, additional, Papa, S, additional, Franzoso, G, additional, and Borsello, T, additional
- Published
- 2006
- Full Text
- View/download PDF
20. Granule neuron DNA damage following deafferentation in adult rats cerebellar cortex: a lesion model
- Author
-
Borsello, T., primary, Di Luzio, A., additional, Ciotti, M.T., additional, Calissano, P., additional, and Galli, C., additional
- Published
- 1999
- Full Text
- View/download PDF
21. Adenosine and ADP prevent apoptosis in cultured rat cerebellar granule cells
- Author
-
Vitolo, O.V., primary, Ciotti, M.T., additional, Galli, C., additional, Borsello, T., additional, and Calissano, P., additional
- Published
- 1998
- Full Text
- View/download PDF
22. Regressive modifications of climbing fibres following purkinje cell degeneration in the cerebellar cortex of the adult rat
- Author
-
Rossi, F., primary, Borsello, T., additional, Vaudano, E., additional, and Strata, P., additional
- Published
- 1993
- Full Text
- View/download PDF
23. 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
- Published
- 2011
24. Blocking ADAM10 synaptic trafficking generates a model of sporadic Alzheimer's disease.
- Author
-
Epis R, Marcello E, Gardoni F, Vastagh C, Malinverno M, Balducci C, Colombo A, Borroni B, Vara H, Dell'agli M, Cattabeni F, Giustetto M, Borsello T, Forloni G, Padovani A, Di Luca M, Epis, Roberta, Marcello, Elena, Gardoni, Fabrizio, and Vastagh, Csaba
- Abstract
We describe here an innovative, non-transgenic animal model of Alzheimer's disease. This model mimics early stages of sporadic disease, which represents the vast majority of cases. The model was obtained by interfering with the complex between a disintegrin and metalloproteinase domain containing protein 10 (ADAM10), the main α-secretase candidate, and its partner, synapse-associated protein 97, a protein of the postsynaptic density-membrane associated guanylate kinase family. Association of ADAM10 with synapse-associated protein 97 governs enzyme trafficking and activity at synapses. Interfering with the ADAM10/synapse-associated protein 97 complex for 2 weeks by means of a cell-permeable peptide strategy is sufficient to shift the metabolism of the amyloid precursor protein towards amyloidogenesis and allows the reproduction of initial phases of sporadic Alzheimer's disease. After 2 weeks of treatment, we detected progressive Alzheimer's disease-like neuropathology, with an increase of β-amyloid aggregate production and of tau hyperphosphorylation, and a selective alteration of N-methyl-d-aspartic acid receptor subunit composition in the postsynaptic compartment of mouse brain. Behavioural and electrophysiological deficits were also induced by peptide treatment. [ABSTRACT FROM AUTHOR]
- Published
- 2010
- Full Text
- View/download PDF
25. Role of the JNK pathway in NMDA-mediated excitotoxicity of cortical neurons.
- Author
-
Centeno, C., Repici, M., Chatton, J.-Y., Riederer, B. M., Bonny, C., Nicod, P., Price, M., Clarke, P. G. H., Papa, S., Franzoso, G., and Borsello, T.
- Subjects
JNK mitogen-activated protein kinases ,NEURODEGENERATION ,METHYL aspartate ,PHOSPHORYLATION ,CEREBRAL ischemia ,PEPTIDES ,EXCITATORY amino acids ,CELLULAR control mechanisms - Abstract
Excitotoxic insults induce c-Jun N-terminal kinase (JNK) activation, which leads to neuronal death and contributes to many neurological conditions such as cerebral ischemia and neurodegenerative disorders. The action of JNK can be inhibited by the D-retro-inverso form of JNK inhibitor peptide (D-JNKI1), which totally prevents death induced by N-methyl-D-aspartate (NMDA) in vitro and strongly protects against different in vivo paradigms of excitotoxicity. To obtain optimal neuroprotection, it is imperative to elucidate the prosurvival action of D-JNKI1 and the death pathways that it inhibits. In cortical neuronal cultures, we first investigate the pathways by which NMDA induces JNK activation and show a rapid and selective phosphorylation of mitogen-activated protein kinase kinase 7 (MKK7), whereas the only other known JNK activator, mitogen-activated protein kinase kinase 4 (MKK4), was unaffected. We then analyze the action of D-JNKI1 on four JNK targets containing a JNK-binding domain: MAPK-activating death domain-containing protein/differentially expressed in normal and neoplastic cells (MADD/DENN), MKK7, MKK4 and JNK-interacting protein-1 (IB1/JIP-1).Cell Death and Differentiation (2007) 14, 240–253. doi:10.1038/sj.cdd.4401988; published online 23 June 2006 [ABSTRACT FROM AUTHOR]
- Published
- 2007
- Full Text
- View/download PDF
26. C-JUN NH2-TERMINAL KINASE MEDIATES THE HEPATIC STRESS RESPONSE AFTER RAT HEMORRHAGE/RESUSCITATION.
- Author
-
Relja, B., Schwestka, B., Sun-Young Lee, V., Henrich, D., Czerny, C., Borsello, T., Lehnert, M., and Marzi, I.
- Published
- 2006
- Full Text
- View/download PDF
27. Super-resolution study of PIAS SUMO E3-ligases in hippocampal and cortical neurons
- Author
-
Tiziana Borsello, Luca Colnaghi, Clara Alice Musi, Luca Russo, Andrea Conz, Conz, A., Musi, C. A., Russo, L., Borsello, T., and Colnaghi, L.
- Subjects
Gene isoform ,Histology ,hippocampus ,QH301-705.5 ,Ubiquitin-Protein Ligases ,hipppocampus ,Biophysics ,SUMO protein ,Regulator ,neurons ,Hippocampal formation ,Biology ,Hippocampus ,Article ,Synapse ,Mice ,synapse ,Animals ,Premovement neuronal activity ,Protein inhibitor of activated STAT ,Biology (General) ,Cerebral Cortex ,Neurons ,Microscopy ,Sumoylation ,Cell Biology ,Protein Inhibitors of Activated STAT ,Cell biology ,cortex ,SUMO ,Synaptic plasticity ,Cortex - Abstract
The SUMOylation machinery is a regulator of neuronal activity and synaptic plasticity. It is composed of SUMO isoforms and specialized enzymes named E1, E2 and E3 SUMO ligases. Recent studies have highlighted how SUMO isoforms and E2 enzymes localize with synaptic markers to support previous functional studies but less information is available on E3 ligases. PIAS proteins - belonging to the protein inhibitor of activated STAT (PIAS) SUMO E3-ligase family - are the best-characterized SUMO E3-ligases and have been linked to the formation of spatial memory in rodents. Whether however they exert their function co-localizing with synaptic markers is still unclear. In this study, we applied for the first time structured illumination microscopy (SIM) to PIAS ligases to investigate the co-localization of PIAS1 and PIAS3 with synaptic markers in hippocampal and cortical murine neurons. The results indicate partial co-localization of PIAS1 and PIAS3 with synaptic markers in hippocampal neurons and much rarer occurrence in cortical neurons. This is in line with previous super-resolution reports describing the co-localization with synaptic markers of other components of the SUMOylation machinery.
- Published
- 2021
28. Deregulated Local Protein Synthesis in the Brain Synaptosomes of a Mouse Model for Alzheimer’s Disease
- Author
-
Luisa Cigliano, Carolina Cefaliello, Eduardo Penna, Antonio Giuditta, Carmela Barbato, Jong Tai Chun, Maria Concetta Miniaci, Carla Perrone-Capano, Giovanna Trinchese, Giuseppina Di Ruberto, Tiziana Borsello, Marianna Crispino, Maria Pina Mollica, Cefaliello, C., Penna, E., Barbato, C., Di Ruberto, G., Mollica, M. P., Trinchese, G., Cigliano, L., Borsello, T., Chun, J. T., Giuditta, A., Perrone Capano, C., Miniaci, M., and Crispino, M.
- Subjects
0301 basic medicine ,Mutant ,Cell ,Neuroscience (miscellaneous) ,Mice, Transgenic ,Plaque, Amyloid ,Inflammation ,Biology ,Synaptic plasticity ,Amyloid beta-Protein Precursor ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,0302 clinical medicine ,Alzheimer Disease ,Neuroplasticity ,Amyloid precursor protein ,medicine ,Protein biosynthesis ,Learning ,Animals ,Neurons ,Memory Disorders ,Amyloid beta-Peptides ,Local protein synthesi ,Brain ,Translation (biology) ,Disease Models, Animal ,030104 developmental biology ,medicine.anatomical_structure ,Neurology ,Synapses ,biology.protein ,medicine.symptom ,Alzheimer’s disease ,Neuroscience ,030217 neurology & neurosurgery ,Synaptosomes - Abstract
While protein synthesis in neurons is largely attributed to cell body and dendrites, the capability of synaptic regions to synthesize new proteins independently of the cell body has been widely demonstrated as an advantageous mechanism subserving synaptic plasticity. Thus, the contribution that local protein synthesis at synapses makes to physiology and pathology of brain plasticity may be more prevalent than initially thought. In this study, we tested if local protein synthesis at synapses is deregulated in the brains of TgCRND8 mice, an animal model for Alzheimer’s disease (AD) overexpressing mutant human amyloid precursor protein (APP). To this end, we used synaptosomes as a model system to study the functionality of the synaptic regions in mouse brains. Our results showed that, while TgCRND8 mice exhibit early signs of brain inflammation and deficits in learning, the electrophoretic profile of newly synthesized proteins in their synaptosomes was subtly different from that of the control mice. Interestingly, APP itself was, in part, locally synthesized in the synaptosomes, underscoring the potential importance of local translation at synapses. More importantly, after the contextual fear conditioning, de novo synthesis of some individual proteins was significantly enhanced in the synaptosomes of control animals, but the TgCRND8 mice failed to display such synaptic modulation by training. Taken together, our results demonstrate that synaptic synthesis of proteins is impaired in the brain of a mouse model for AD, and raise the possibility that this deregulation may contribute to the early progression of the pathology.
- Published
- 2019
- Full Text
- View/download PDF
29. Neuronal Localization of SENP Proteins with Super Resolution Microscopy
- Author
-
Luana Fioriti, Luca Colnaghi, Luca Russo, Mario Salmona, Andrea Conz, Tiziana Borsello, Clara Alice Musi, Colnaghi, L, Conz, A, Russo, L, Musi, Ca, Fioriti, L, Borsello, T, and Salmona
- Subjects
Protein sumoylation ,SENP1 ,medicine.medical_treatment ,SENP6 ,SUMO protein ,neurons ,Article ,lcsh:RC321-571 ,03 medical and health sciences ,0302 clinical medicine ,SENPs ,medicine ,lcsh:Neurosciences. Biological psychiatry. Neuropsychiatry ,030304 developmental biology ,0303 health sciences ,Protease ,biology ,Chemistry ,General Neuroscience ,Colocalization ,Cysteine protease ,Cell biology ,SUMO ,super resolution microscopy ,Synaptophysin ,biology.protein ,030217 neurology & neurosurgery - Abstract
SUMOylation of proteins plays a key role in modulating neuronal function. For this reason, the balance between protein SUMOylation and deSUMOylation requires fine regulation to guarantee the homeostasis of neural tissue. While extensive research has been carried out on the localization and function of small ubiquitin-related modifier (SUMO) variants in neurons, less attention has been paid to the SUMO-specific isopeptidases that constitute the human SUMO-specific isopeptidase (SENP)/Ubiquitin-Specific Protease (ULP) cysteine protease family (SENP1-3 and SENP5-7). Here, for the first time, we studied the localization of SENP1, SENP6, and SENP7 in cultured hippocampal primary neurons at a super resolution detail level, with structured illumination microscopy (SIM). We found that the deSUMOylases partially colocalize with pre- and post-synaptic markers such as synaptophysin and drebrin. Thus, further confirming the presence with synaptic markers of the negative regulators of the SUMOylation machinery.
- Published
- 2020
30. A New Fluorogenic Peptide Determines Proteasome Activity in Single Cells
- Author
-
Rodolfo Gonella Diaza, Mario Salmona, Simone Cenci, Enrico Monzani, Paolo Cascio, Roberto Sitia, Tiziana Borsello, Enrico Davoli, Pietro Veglianese, Niccolò Pengo, Ada De Luigi, Eugenio Erba, Andrea Carrà, Silvana Anna Maria Urru, Elena Fumagalli, Gianluigi Forloni, URRU S. A., M, Veglianese, P, DE LUIGI, A, Fumagalli, E, Erba, E, GONELLA DIAZA, R, Carra, A, Davoli, E, Borsello, T, Forloni, G, Pengo, N, Monzani, E, Cascio, P, Cenci, S, Sitia, R, and Salmona, M
- Subjects
Models, Molecular ,Proteasome Endopeptidase Complex ,Cell ,Proteasome ,Protein degradation ,Proteasomal activity ,TAT ,Peptide ,Hippocampus ,Flow cytometry ,Mice ,chemistry.chemical_compound ,In vivo ,Cell Line, Tumor ,Drug Discovery ,medicine ,Animals ,Humans ,Fluorescent Dyes ,Neurons ,chemistry.chemical_classification ,medicine.diagnostic_test ,Chemistry ,Flow Cytometry ,In vitro ,Cell biology ,Protein Subunits ,medicine.anatomical_structure ,Microscopy, Fluorescence ,Biochemistry ,EDANS ,Molecular Medicine ,Peptides - Abstract
The ubiquitin−proteasome system plays a critical role in many diseases, making it an attractive biomarker and therapeutic target. However, the impact of results obtained in vitro using purified proteasome particles or whole cell extracts is limited by the lack of efficient methods to assess proteasome activity in living cells. We have engineered an internally quenched fluorogenic peptide with a proteasome-specific cleavage motif fused to TAT and linked to the fluorophores DABCYL and EDANS. This peptide penetrates cell membranes and is rapidly cleaved by the proteasomal chymotrypsin-like activity, generating a quantitative fluorescent reporter of in vivo proteasome activity as assessed by time-lapse or flow cytometry fluorescence analysis. This reporter is an innovative tool for monitoring proteasomal proteolytic activities in physiological and pathological conditions.
- Published
- 2010
- Full Text
- View/download PDF
31. The cell-permeable Aβ1-6A2VTAT(D) peptide reverts synaptopathy induced by Aβ1-42wt
- Author
-
Mario Salmona, Giuseppe Di Fede, Fabrizio Tagliavini, Sara Cimini, Laura Colombo, Alfredo Cagnotto, Simona Mancini, Alessandra Sclip, Tiziana Borsello, Massimo Messa, Cimini, S, Sclip, A, Mancini, S, Colombo, L, Messa, M, Cagnotto, A, Di Fede, G, Tagliavini, F, Salmona, M, and Borsello, T
- Subjects
0301 basic medicine ,Cell Membrane Permeability ,Aβ oligomers ,Synaptic injury ,Aβ oligomer ,Dendritic Spines ,Peptide ,Mice, Transgenic ,Biology ,medicine.disease_cause ,Neuroprotection ,Hippocampus ,lcsh:RC321-571 ,03 medical and health sciences ,Mice ,In vivo ,Alzheimer Disease ,Brainbow hippocampal neurons ,medicine ,Animals ,lcsh:Neurosciences. Biological psychiatry. Neuropsychiatry ,chemistry.chemical_classification ,Neurons ,Mutation ,Cell-permeable peptide ,Amyloid beta-Peptides ,Wild type ,Long-term potentiation ,Alzheimer's disease ,medicine.disease ,Peptide Fragments ,Cell biology ,Disease Models, Animal ,030104 developmental biology ,Biochemistry ,chemistry ,Brainbow hippocampal neuron ,Neurology ,Synapses ,Synaptopathy ,A673V mutation - 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.
- Published
- 2015
32. Soluble Aβ oligomer-induced synaptopathy: c-Jun N-terminal kinase's role
- Author
-
Laura Colombo, Egbert Welker, Federica Morelli, Simona Mancini, Andrea Arnaboldi, Pietro Veglianese, Isabella Colombo, Tiziana Borsello, Mario Salmona, Massimo Messa, Alessandra Sclip, Xanthi Antoniou, Sara Cimini, 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
Dendritic spine ,Dendritic Spines ,Caspase 3 ,Alzhreimer's disease ,Mitogen-activated protein kinase kinase ,Models, Biological ,Receptors, N-Methyl-D-Aspartate ,Oligomer ,Mice ,chemistry.chemical_compound ,Alzheimer Disease ,Genetics ,medicine ,Animals ,Humans ,Receptors, AMPA ,Molecular Biology ,Amyloid beta-Peptides ,Chemistry ,Kinase ,c-jun ,JNK Mitogen-Activated Protein Kinases ,Cell Biology ,General Medicine ,medicine.disease ,c-Jun N-terminal kinase, JNK, Amyloid-beta oligomers ,Cell biology ,Synaptopathy ,Signal transduction ,Signal Transduction - Published
- 2013
- Full Text
- View/download PDF
33. A new APP mutation prevents synaptic degeneration in Alzheimer Disease model
- Author
-
Mario Salmona, Alessandro Rossi, Alessandra Sclip, G. Di Fede, Fabrizio Tagliavini, Laura Colombo, Simona Mancini, Tiziana Borsello, Massimo Messa, Borsello, T, Sclip, A, Mancini, S, Colombo, L, Rossi, A, Messa, M, Di Fede, G, Tagliavini, F, and Salmona, M
- Subjects
Aging ,business.industry ,General Neuroscience ,Aβ oligomers ,Alzheimer's Disease, Aβ1-42, A673V mutation, Aβ oligomers ,Degeneration (medical) ,medicine.disease ,Mutation (genetic algorithm) ,Cancer research ,Medicine ,Neurology (clinical) ,Geriatrics and Gerontology ,Alzheimer's disease ,business ,Developmental Biology - Published
- 2014
- Full Text
- View/download PDF
34. The Stem Cells as a Potential Treatment for Neurodegeneration
- Author
-
Daniele Bottai, Ferrari Daniela, Angelo L. Vescovi, Borsello, T, Ferrari, D, Vescovi, A, and Bottai, D
- Subjects
Male ,Time Factor ,Central nervous system ,Cell ,Cell Separation ,Biology ,Mice ,Stem Cell ,medicine ,Spinal cord injury ,Transplantation, Homologou ,Neurodegenerative Disease ,Animal ,Multiple sclerosis ,Neurodegeneration ,BIO/13 - BIOLOGIA APPLICATA ,Neuron ,medicine.disease ,Neural stem cell ,Transplantation ,Disease Models, Animal ,medicine.anatomical_structure ,Female ,Stem cell ,Cell Culture Technique ,Neuroscience ,Human ,Stem Cell Transplantation - Abstract
Cell degeneration and death, be it extensive and widespread, such as in metabolic disorders, or focal and selective as in Parkinson's disease (PD), is the underlying feature of many neurological diseases. Thus, the replacement of cells lost by injury or disease has become a central tenet in strategies aiming at the development of novel therapeutic approaches for neurodegenerative disorders. In addition to the in vivo recruitment of endogenous cells, which is now emerging as a promising novel strategy, the transplantation of new, exogenously generated brain cells is probably the most extensively studied methodology for cell replacement in the central nervous system, with the initial experimental clinical studies in PD dating back to the early 1970s (Bjorklund, A. and Stenevi, U., 1984, Intracerebral neural implants: neuronal replacement and reconstruction of damaged circuitries. Annu Rev Neurosci 7, 279-308; Snyder, B. J. and Olanow, C. W., 2005, Stem cell treatment for Parkinson's disease: an update for 2005. Curr Opin Neurol 18, 376-85). The need to generate the cells to be transplanted in large quantities and in a reproducible, steady, and safe fashion has long represented one of the major issues in this field, regardless of whether one was trying to produce specific cell subtypes or uncommitted and highly plastic neural precursors, which would respond to local, instructive cues, upon transplantation into the damaged area. Neural stem cells (NSCs), with their capacity for long-term expansion in vitro and their extensive functional stability and plasticity, allow now for the establishment of cultures of mature neural cells as well as highly undifferentiated precursors and are emerging as one of the most amenable cell sources for neural transplantation (Gage, F. H., 2000, Mammalian neural stem cells. Science 287, 1433-8; McKay, R., 1997, Stem cells in the central nervous system. Science 276, 66-71). This chapter illustrates the basic aspect of the handling and preparation of NSCs for experimental transplantation in two animal models of neurodegenerative disorders, namely, postcontusion spinal cord injury and multiple sclerosis.
- Published
- 2007
- Full Text
- View/download PDF
35. Inflammation and depression: A study protocol to dissect pathogenetic mechanisms in the onset, comorbidity and treatment response.
- Author
-
Scassellati C, Cattane N, Benedetti F, Borsello T, Cicala G, Gennarelli M, Genini P, Gialluisi A, Giani A, Iacoviello L, Minelli A, Spina E, Vai B, Vitali E, and Cattaneo A
- Abstract
About one third of patients suffering from Major Depressive Disorder (MDD) do not respond to any antidepressant medications and 75% experience relapses and general health deterioration. Importantly, inflammation can contribute to such negative outcomes, as well as to cause depression in patients who have been exposed to adverse childhood experiences and/or to viral infections, including COVID-19. Depressed patients also have an increased risk for developing comorbidities, such as cardio-metabolic dysfunctions, where inflammatory alterations, again, play a role in connecting MDD and these comorbid conditions. Here, we present our study protocol funded by the Italian Ministry of Health in the context of the PNRR call (M6/C2_CALL 2022; Project code: PNRR-MAD-2022-12375859). The project aims to clarify the role of inflammation: i) in the onset of depression in association with environmental factors; ii) in the mechanisms associated with treatment response/resistance; iii) in depression and its comorbidity. To reach all these aims, we will perform biochemical, transcriptomic, genetic variants analyses on inflammatory/immune genes, pharmacokinetics and machine learning techniques, taking advantage of different human cohorts (adolescent depressed patients exposed to childhood trauma; adult depressed patients; treatment resistant depression patients; both prevalent and incident depression cases identified within a large population cohort). Moreover, we will use in vitro models (primary cultures of astrocytes, neurons and microglia) treated with pro-inflammatory or stressful challenges and preventive compounds to clarify the underlying mechanisms. This 2-years project will increase the knowledge on the role of inflammation in the prevention and treatment of MDD and in comorbid disorders, and it will also provide experimental evidence for the development of novel targets and tools for innovative personalized intervention strategies., Competing Interests: none., (© 2024 Published by Elsevier Inc.)
- Published
- 2024
- Full Text
- View/download PDF
36. JNK Activation Correlates with Cognitive Impairment and Alteration of the Post-Synaptic Element in the 5xFAD AD Mouse Model.
- Author
-
Priori EC, Musi CA, Giani A, Colnaghi L, Milic I, Devitt A, Borsello T, and Repici M
- Subjects
- Animals, Mice, MAP Kinase Signaling System, Phosphorylation, Disease Models, Animal, Alzheimer Disease metabolism, Cognitive Dysfunction metabolism, JNK Mitogen-Activated Protein Kinases metabolism
- Abstract
The c-Jun N-terminal kinases (JNKs) are a family of proteins that, once activated by stress stimuli, can alter neuronal functions and survival. The JNK cascade plays a crucial role in the post-synaptic neuronal compartment by altering its structural organization and leading, at worst, to an overall impairment of neuronal communication. Increasing evidence suggests that synaptic impairment is the first neurodegenerative event in Alzheimer's disease (AD). To better elucidate this mechanism, we longitudinally studied 5xFAD mice at three selected time points representative of human AD symptom progression. We tested the mice cognitive performance by using the radial arm water maze (RAWM) in parallel with biochemical evaluations of post-synaptic enriched protein fraction and total cortical parenchyma. We found that 5xFAD mice presented a strong JNK activation at 3.5 months of age in the post-synaptic enriched protein fraction. This JNK activation correlates with a structural alteration of the post-synaptic density area and with memory impairment at this early stage of the disease that progressively declines to cause cell death. These findings pave the way for future studies on JNK as a key player in early neurodegeneration and as an important therapeutic target for the development of new compounds able to tackle synaptic impairment in the early phase of AD pathology.
- Published
- 2023
- Full Text
- View/download PDF
37. Synaptic alterations as a common phase in neurological and neurodevelopmental diseases: JNK is a key mediator in synaptic changes.
- Author
-
Musi CA, Bonadonna C, and Borsello T
- Abstract
Competing Interests: None
- Published
- 2023
- Full Text
- View/download PDF
38. Salivary microRNA profiling dysregulation in autism spectrum disorder: A pilot study.
- Author
-
Kalemaj Z, Marino MM, Santini AC, Tomaselli G, Auti A, Cagetti MG, Borsello T, Costantino A, Inchingolo F, Boccellino M, Di Domenico M, and Tartaglia GM
- Abstract
Introduction: Autism spectrum disorders (ASD) are the most prevalent neurobiological disorders in children. The etiology comprises genetic, epigenetic, and environmental factors such as dysfunction of the immune system. Epigenetic mechanisms are mainly represented by DNA methylation, histone modifications, and microRNAs (miRNA). The major explored epigenetic mechanism is mediated by miRNAs which target genes known to be involved in ASD pathogenesis. Salivary poly-omic RNA measurements have been associated with ASD and are helpful to differentiate ASD endophenotypes. This study aims to comprehensively examine miRNA expression in children with ASD and to reveal potential biomarkers and possible disease mechanisms so that they can be used to improve faction between individuals by promoting more personalized therapeutic approaches., Materials and Methods: Saliva samples were collected from 10 subjects: 5 samples of children with ASD and 5 from healthy controls. miRNAs were analyzed using an Illumina Next-Generation-Sequencing (NGS) system., Results: Preliminary data highlighted the presence of 365 differentially expressed miRNAs. Pathway analysis, molecular function, biological processes, and target genes of 41 dysregulated miRNAs were assessed, of which 20 were upregulated, and 21 were downregulated in children with ASD compared to healthy controls., Conclusion: The results of this study represent preliminary but promising data, as the identified miRNA pathways could represent useful biomarkers for the early non-invasive diagnosis of ASD., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2022 Kalemaj, Marino, Santini, Tomaselli, Auti, Cagetti, Borsello, Costantino, Inchingolo, Boccellino, Di Domenico and Tartaglia.)
- Published
- 2022
- Full Text
- View/download PDF
39. Effect of 3D Synthetic Microscaffold Nichoid on the Morphology of Cultured Hippocampal Neurons and Astrocytes.
- Author
-
Musi CA, Colnaghi L, Giani A, Priori EC, Marchini G, Tironi M, Conci C, Cerullo G, Osellame R, Raimondi MT, Remuzzi A, and Borsello T
- Subjects
- Coculture Techniques, Hippocampus, Humans, Neurons metabolism, Astrocytes, Brain Diseases metabolism
- Abstract
The human brain is the most complex organ in biology. This complexity is due to the number and the intricate connections of brain cells and has so far limited the development of in vitro models for basic and applied brain research. We decided to create a new, reliable, and cost-effective in vitro system based on the Nichoid, a 3D microscaffold microfabricated by two-photon laser polymerization technology. We investigated whether these 3D microscaffold devices can create an environment allowing the manipulation, monitoring, and functional assessment of a mixed population of brain cells in vitro. With this aim, we set up a new model of hippocampal neurons and astrocytes co-cultured in the Nichoid microscaffold to generate brain micro-tissues of 30 μm thickness. After 21 days in culture, we morphologically characterized the 3D spatial organization of the hippocampal astrocytes and neurons within the microscaffold, and we compared our observations to those made using the classical 2D co-culture system. We found that the co-cultured cells colonized the entire volume of the 3D devices. Using confocal microscopy, we observed that within this period the different cell types had become well-differentiated. This was further elaborated with the use of drebrin, PSD-95, and synaptophysin antibodies that labeled the majority of neurons, both in the 2D as well as in the 3D co-cultures. Using scanning electron microscopy, we found that neurons in the 3D co-culture displayed a significantly larger amount of dendritic protrusions compared to neurons in the 2D co-culture. This latter observation indicates that neurons growing in a 3D environment may be more prone to form connections than those co-cultured in a 2D condition. Our results show that the Nichoid can be used as a 3D device to investigate the structure and morphology of neurons and astrocytes in vitro. In the future, this model can be used as a tool to study brain cell interactions in the discovery of important mechanisms governing neuronal plasticity and to determine the factors that form the basis of different human brain diseases. This system may potentially be further used for drug screening in the context of various brain diseases.
- Published
- 2022
- Full Text
- View/download PDF
40. Colocalization and Interaction Study of Neuronal JNK3, JIP1, and β-Arrestin2 Together with PSD95.
- Author
-
Musi CA, Marchini G, Giani A, Tomaselli G, Priori EC, Colnaghi L, and Borsello T
- Subjects
- Disks Large Homolog 4 Protein metabolism, Humans, JNK Mitogen-Activated Protein Kinases metabolism, Male, Neurons metabolism, Phosphorylation, beta-Arrestin 1, Mitogen-Activated Protein Kinase 10 metabolism, Mitogen-Activated Protein Kinases metabolism
- Abstract
c-Jun N-terminal kinases (JNKs) are stress-activated serine/threonine protein kinases belonging to the mitogen-activated protein kinase (MAPK) family. Among them, JNK3 is selectively expressed in the central nervous system, cardiac smooth muscle, and testis. In addition, it is the most responsive JNK isoform to stress stimuli in the brain, and it is involved in synaptic dysfunction, an essential step in neurodegenerative processes. JNK3 pathway is organized in a cascade of amplification in which signal transduction occurs by stepwise, highly controlled phosphorylation. Since different MAPKs share common upstream activators, pathway specificity is guaranteed by scaffold proteins such as JIP1 and β-arrestin2. To better elucidate the physiological mechanisms regulating JNK3 in neurons, and how these interactions may be involved in synaptic (dys)function, we used (i) super-resolution microscopy to demonstrate the colocalization among JNK3-PSD95-JIP1 and JNK3-PSD95-β-arrestin2 in cultured hippocampal neurons, and (ii) co-immunoprecipitation techniques to show that the two scaffold proteins and JNK3 can be found interacting together with PSD95. The protein-protein interactions that govern the formation of these two complexes, JNK3-PSD95-JIP1 and JNK3-PSD95-β-arrestin2, may be used as targets to interfere with their downstream synaptic events.
- Published
- 2022
- Full Text
- View/download PDF
41. Common pathways in dementia and diabetic retinopathy: understanding the mechanisms of diabetes-related cognitive decline.
- Author
-
Little K, Llorián-Salvador M, Scullion S, Hernández C, Simó-Servat O, Del Marco A, Bosma E, Vargas-Soria M, Carranza-Naval MJ, Van Bergen T, Galbiati S, Viganò I, Musi CA, Schlingemann R, Feyen J, Borsello T, Zerbini G, Klaassen I, Garcia-Alloza M, Simó R, and Stitt AW
- Subjects
- Glycation End Products, Advanced metabolism, Humans, Alzheimer Disease metabolism, Cognitive Dysfunction etiology, Diabetes Mellitus, Type 2 complications, Diabetic Retinopathy metabolism, Diabetic Retinopathy pathology
- Abstract
Type 2 diabetes (T2D) is associated with multiple comorbidities, including diabetic retinopathy (DR) and cognitive decline, and T2D patients have a significantly higher risk of developing Alzheimer's disease (AD). Both DR and AD are characterized by a number of pathological mechanisms that coalesce around the neurovascular unit, including neuroinflammation and degeneration, vascular degeneration, and glial activation. Chronic hyperglycemia and insulin resistance also play a significant role, leading to activation of pathological mechanisms such as increased oxidative stress and the accumulation of advanced glycation end-products (AGEs). Understanding these common pathways and the degree to which they occur simultaneously in the brain and retina during diabetes will provide avenues to identify T2D patients at risk of cognitive decline., Competing Interests: Declaration of interests No interests are declared., (Copyright © 2021. Published by Elsevier Ltd.)
- Published
- 2022
- Full Text
- View/download PDF
42. Synaptic Proteins as Fluid Biomarkers in Alzheimer's Disease: A Systematic Review and Meta-Analysis.
- Author
-
Roveta F, Cermelli A, Boschi S, Ferrandes F, Grassini A, Marcinnò A, Spina M, Rubino E, Borsello T, Vercelli A, and Rainero I
- Subjects
- Humans, GAP-43 Protein, Neurogranin cerebrospinal fluid, Biomarkers cerebrospinal fluid, tau Proteins cerebrospinal fluid, Amyloid beta-Peptides cerebrospinal fluid, Alzheimer Disease diagnosis, Alzheimer Disease cerebrospinal fluid, Cognitive Dysfunction diagnosis
- Abstract
Background: Synaptic disruption precedes neuronal death and correlates with clinical features of Alzheimer's disease (AD). The identification of fluid biomarkers of synaptic damage is emerging as a goal for early and accurate diagnosis of the disease., Objective: To perform a systematic review and meta-analysis to determine whether fluid biomarkers of synaptic damage are impaired in AD., Methods: PubMed, Scopus, EMBASE, and Web of Science were searched for articles reporting synaptic proteins as fluid biomarkers in AD and cognitively unimpaired (CU) individuals. Pooled effect sizes were determined using the Hedge G method with random effects. Questions adapted from the Quality Assessment of Diagnostic Accuracy Studies were applied for quality assessment. A protocol for this study has been previously registered in PROSPERO (registration number: CRD42021277487)., Results: The search strategy identified 204 articles that were assessed for eligibility. A total of 23 studies were included in the systematic review and 15 were included in the meta-analysis. For Neurogranin, 827 AD and 1,237 CU subjects were included in the meta-analysis, showing a significant increase in cerebrospinal fluid of patients with AD compared to CU individuals, with an effect size of 1.01 (p < 0.001). A significant increase in SNAP-25 and GAP-43 levels in CSF of patients with AD was observed., Conclusion: Neurogranin, SNAP-25, and GAP-43 are possible biomarkers of synaptic damage in AD, and other potential synaptic biomarkers are emerging. This meta-analysis also revealed that there are still relatively few studies investigating these biomarkers in patients with AD or other dementias and showed wide heterogeneity in literature.
- Published
- 2022
- Full Text
- View/download PDF
43. JNK signaling provides a novel therapeutic target for Rett syndrome.
- Author
-
Musi CA, Castaldo AM, Valsecchi AE, Cimini S, Morello N, Pizzo R, Renieri A, Meloni I, Bonati M, Giustetto M, and Borsello T
- Subjects
- Animals, Disease Models, Animal, MAP Kinase Signaling System, Mice, Neurons metabolism, Synapses metabolism, Rett Syndrome genetics, Rett Syndrome metabolism, Rett Syndrome therapy
- Abstract
Background: Rett syndrome (RTT) is a monogenic X-linked neurodevelopmental disorder characterized by loss-of-function mutations in the MECP2 gene, which lead to structural and functional changes in synapse communication, and impairments of neural activity at the basis of cognitive deficits that progress from an early age. While the restoration of MECP2 in animal models has been shown to rescue some RTT symptoms, gene therapy intervention presents potential side effects, and with gene- and RNA-editing approaches still far from clinical application, strategies focusing on signaling pathways downstream of MeCP2 may provide alternatives for the development of more effective therapies in vivo. Here, we investigate the role of the c-Jun N-terminal kinase (JNK) stress pathway in the pathogenesis of RTT using different animal and cell models and evaluate JNK inhibition as a potential therapeutic approach., Results: We discovered that the c-Jun N-terminal kinase (JNK) stress pathway is activated in Mecp2-knockout, Mecp2-heterozygous mice, and in human MECP2-mutated iPSC neurons. The specific JNK inhibitor, D-JNKI1, promotes recovery of body weight and locomotor impairments in two mouse models of RTT and rescues their dendritic spine alterations. Mecp2-knockout presents intermittent crises of apnea/hypopnea, one of the most invalidating RTT pathological symptoms, and D-JNKI1 powerfully reduces this breathing dysfunction. Importantly, we discovered that also neurons derived from hiPSC-MECP2 mut show JNK activation, high-phosphorylated c-Jun levels, and cell death, which is not observed in the isogenic control wt allele hiPSCs. Treatment with D-JNKI1 inhibits neuronal death induced by MECP2 mutation in hiPSCs mut neurons., Conclusions: As a summary, we found altered JNK signaling in models of RTT and suggest that D-JNKI1 treatment prevents clinical symptoms, with coherent results at the cellular, molecular, and functional levels. This is the first proof of concept that JNK plays a key role in RTT and its specific inhibition offers a new and potential therapeutic tool to tackle RTT., (© 2021. The Author(s).)
- Published
- 2021
- Full Text
- View/download PDF
44. Super-resolution study of PIAS SUMO E3-ligases in hippocampal and cortical neurons.
- Author
-
Conz A, Musi CA, Russo L, Borsello T, and Colnaghi L
- Subjects
- Animals, Cerebral Cortex ultrastructure, Hippocampus ultrastructure, Mice, Neurons ultrastructure, Cerebral Cortex enzymology, Hippocampus enzymology, Microscopy methods, Neurons enzymology, Protein Inhibitors of Activated STAT metabolism, Sumoylation, Ubiquitin-Protein Ligases metabolism
- Abstract
The SUMOylation machinery is a regulator of neuronal activity and synaptic plasticity. It is composed of SUMO isoforms and specialized enzymes named E1, E2 and E3 SUMO ligases. Recent studies have highlighted how SUMO isoforms and E2 enzymes localize with synaptic markers to support previous functional studies but less information is available on E3 ligases. PIAS proteins - belonging to the protein inhibitor of activated STAT (PIAS) SUMO E3-ligase family - are the best-characterized SUMO E3-ligases and have been linked to the formation of spatial memory in rodents. Whether however they exert their function co-localizing with synaptic markers is still unclear. In this study, we applied for the first time structured illumination microscopy (SIM) to PIAS ligases to investigate the co-localization of PIAS1 and PIAS3 with synaptic markers in hippocampal and cortical murine neurons. The results indicate partial co-localization of PIAS1 and PIAS3 with synaptic markers in hippocampal neurons and much rarer occurrence in cortical neurons. This is in line with previous super-resolution reports describing the co-localization with synaptic markers of other components of the SUMOylation machinery.
- Published
- 2021
- Full Text
- View/download PDF
45. c-Jun N-terminal kinase 1 (JNK1) modulates oligodendrocyte progenitor cell architecture, proliferation and myelination.
- Author
-
Lorenzati M, Boda E, Parolisi R, Bonato M, Borsello T, Herdegen T, Buffo A, and Vercelli A
- Subjects
- Animals, Mice, Mice, Knockout, Mitogen-Activated Protein Kinase 8 genetics, Myelin Sheath genetics, Cell Proliferation, MAP Kinase Signaling System, Mitogen-Activated Protein Kinase 8 metabolism, Myelin Sheath metabolism, Oligodendrocyte Precursor Cells enzymology, Oligodendroglia enzymology
- Abstract
During Central Nervous System ontogenesis, myelinating oligodendrocytes (OLs) arise from highly ramified and proliferative precursors called oligodendrocyte progenitor cells (OPCs). OPC architecture, proliferation and oligodendro-/myelino-genesis are finely regulated by the interplay of cell-intrinsic and extrinsic factors. A variety of extrinsic cues converge on the extracellular signal-regulated kinase/mitogen activated protein kinase (ERK/MAPK) pathway. Here we found that the germinal ablation of the MAPK c-Jun N-Terminal Kinase isoform 1 (JNK1) results in a significant reduction of myelin in the cerebral cortex and corpus callosum at both postnatal and adult stages. Myelin alterations are accompanied by higher OPC density and proliferation during the first weeks of life, consistent with a transient alteration of mechanisms regulating OPC self-renewal and differentiation. JNK1 KO OPCs also show smaller occupancy territories and a less complex branching architecture in vivo. Notably, these latter phenotypes are recapitulated in pure cultures of JNK1 KO OPCs and of WT OPCs treated with the JNK inhibitor D-JNKI-1. Moreover, JNK1 KO and WT D-JNKI-1 treated OLs, while not showing overt alterations of differentiation in vitro, display a reduced surface compared to controls. Our results unveil a novel player in the complex regulation of OPC biology, on the one hand showing that JNK1 ablation cell-autonomously determines alterations of OPC proliferation and branching architecture and, on the other hand, suggesting that JNK1 signaling in OLs participates in myelination in vivo.
- Published
- 2021
- Full Text
- View/download PDF
46. Neuronal Localization of SENP Proteins with Super Resolution Microscopy.
- Author
-
Colnaghi L, Conz A, Russo L, Musi CA, Fioriti L, Borsello T, and Salmona M
- Abstract
SUMOylation of proteins plays a key role in modulating neuronal function. For this reason, the balance between protein SUMOylation and deSUMOylation requires fine regulation to guarantee the homeostasis of neural tissue. While extensive research has been carried out on the localization and function of small ubiquitin-related modifier (SUMO) variants in neurons, less attention has been paid to the SUMO-specific isopeptidases that constitute the human SUMO-specific isopeptidase (SENP)/Ubiquitin-Specific Protease (ULP) cysteine protease family (SENP1-3 and SENP5-7). Here, for the first time, we studied the localization of SENP1, SENP6, and SENP7 in cultured hippocampal primary neurons at a super resolution detail level, with structured illumination microscopy (SIM). We found that the deSUMOylases partially colocalize with pre- and post-synaptic markers such as synaptophysin and drebrin. Thus, further confirming the presence with synaptic markers of the negative regulators of the SUMOylation machinery.
- Published
- 2020
- Full Text
- View/download PDF
47. JNK3 as Therapeutic Target and Biomarker in Neurodegenerative and Neurodevelopmental Brain Diseases.
- Author
-
Musi CA, Agrò G, Santarella F, Iervasi E, and Borsello T
- Subjects
- Biomarkers metabolism, Brain pathology, Humans, MAP Kinase Signaling System genetics, Molecular Targeted Therapy, Neurodegenerative Diseases metabolism, Neurodegenerative Diseases pathology, Neurodevelopmental Disorders metabolism, Neurodevelopmental Disorders pathology, Phosphorylation, Brain metabolism, Mitogen-Activated Protein Kinase 10 genetics, Neurodegenerative Diseases genetics, Neurodevelopmental Disorders genetics
- Abstract
The c-Jun N -terminal kinase 3 (JNK3) is the JNK isoform mainly expressed in the brain. It is the most responsive to many stress stimuli in the central nervous system from ischemia to Aβ oligomers toxicity. JNK3 activity is spatial and temporal organized by its scaffold protein, in particular JIP-1 and β-arrestin-2, which play a crucial role in regulating different cellular functions in different cellular districts. Extensive evidence has highlighted the possibility of exploiting these adaptors to interfere with JNK3 signaling in order to block its action. JNK plays a key role in the first neurodegenerative event, the perturbation of physiological synapse structure and function, known as synaptic dysfunction. Importantly, this is a common mechanism in many different brain pathologies. Synaptic dysfunction and spine loss have been reported to be pharmacologically reversible, opening new therapeutic directions in brain diseases. Being JNK3-detectable at the peripheral level, it could be used as a disease biomarker with the ultimate aim of allowing an early diagnosis of neurodegenerative and neurodevelopment diseases in a still prodromal phase.
- Published
- 2020
- Full Text
- View/download PDF
48. JNK signaling activation in the Ube3a maternal deficient mouse model: its specific inhibition prevents post-synaptic protein-enriched fraction alterations and cognitive deficits in Angelman Syndrome model.
- Author
-
Musi CA, Agrò G, Buccarello L, Camuso S, and Borsello T
- Subjects
- Animals, Cell Nucleus metabolism, Disease Models, Animal, Female, Hippocampus metabolism, Male, Mice, Neurons metabolism, Angelman Syndrome metabolism, Cognitive Dysfunction metabolism, MAP Kinase Signaling System, Synapses metabolism, Ubiquitin-Protein Ligases metabolism
- Abstract
Deficiency of the E3 ubiquitin ligase UBE3A leads to the neurodevelopmental disorder Angelman syndrome (AS), while higher levels are linked to autism spectrum disorder. The mechanisms underlying the downstream effects of UBE3A loss or gain of function in these disorders are still not well understood, and treatments are still lacking. Here, using the Ube3a maternal loss (Ube3a
m-/p+ ) mouse model, we report an important JNK signaling activation in the hippocampus, cortex and cerebellum correlating with the onset of behavioral defects and biochemical marker alterations in the post-synaptic element, suggesting important spine pathology. JNK activation occurs at 7 and persists up till 23 weeks in Ube3am-/p+ mice in two different cellular compartments: the nucleus and the post-synaptic protein-enriched fraction. To study JNK's role in Ube3am-/p+ pathology we treated mice with the specific JNK inhibitor peptide, D-JNKI1, from 7 to 23 weeks of age. Preventing JNK action in vivo restores the post-synaptic protein-enriched fraction defects and the cognitive impairment in these mice. Our results imply a critical role of UBE3A-JNK signaling in the pathogenesis of UBE3A-related disorders. In particular, it was clear that JNK is a key player in regulating AS synaptic alterations and the correlated cognitive impairments, in fact, its specific inhibition tackles Ube3am-/p+ pathology. This study sheds new light on the neuronal functions of UBE3A and offers new prospects for understanding the pathogenesis of UBE3A-related disorders., Competing Interests: Declaration of Competing Interest The authors declare no actual or potential conflicts of interest., (Copyright © 2020 The Authors. Published by Elsevier Inc. All rights reserved.)- Published
- 2020
- Full Text
- View/download PDF
49. Deregulated Local Protein Synthesis in the Brain Synaptosomes of a Mouse Model for Alzheimer's Disease.
- Author
-
Cefaliello C, Penna E, Barbato C, Di Ruberto G, Mollica MP, Trinchese G, Cigliano L, Borsello T, Chun JT, Giuditta A, Perrone-Capano C, Miniaci MC, and Crispino M
- Subjects
- Alzheimer Disease pathology, Amyloid beta-Peptides metabolism, Amyloid beta-Protein Precursor metabolism, Animals, Disease Models, Animal, Memory Disorders metabolism, Mice, Transgenic, Plaque, Amyloid pathology, Synaptosomes metabolism, Alzheimer Disease metabolism, Brain metabolism, Neurons metabolism, Synapses metabolism
- Abstract
While protein synthesis in neurons is largely attributed to cell body and dendrites, the capability of synaptic regions to synthesize new proteins independently of the cell body has been widely demonstrated as an advantageous mechanism subserving synaptic plasticity. Thus, the contribution that local protein synthesis at synapses makes to physiology and pathology of brain plasticity may be more prevalent than initially thought. In this study, we tested if local protein synthesis at synapses is deregulated in the brains of TgCRND8 mice, an animal model for Alzheimer's disease (AD) overexpressing mutant human amyloid precursor protein (APP). To this end, we used synaptosomes as a model system to study the functionality of the synaptic regions in mouse brains. Our results showed that, while TgCRND8 mice exhibit early signs of brain inflammation and deficits in learning, the electrophoretic profile of newly synthesized proteins in their synaptosomes was subtly different from that of the control mice. Interestingly, APP itself was, in part, locally synthesized in the synaptosomes, underscoring the potential importance of local translation at synapses. More importantly, after the contextual fear conditioning, de novo synthesis of some individual proteins was significantly enhanced in the synaptosomes of control animals, but the TgCRND8 mice failed to display such synaptic modulation by training. Taken together, our results demonstrate that synaptic synthesis of proteins is impaired in the brain of a mouse model for AD, and raise the possibility that this deregulation may contribute to the early progression of the pathology.
- Published
- 2020
- Full Text
- View/download PDF
50. The Parkinson's Disease-Linked Protein DJ-1 Associates with Cytoplasmic mRNP Granules During Stress and Neurodegeneration.
- Author
-
Repici M, Hassanjani M, Maddison DC, Garção P, Cimini S, Patel B, Szegö ÉM, Straatman KR, Lilley KS, Borsello T, Outeiro TF, Panman L, and Giorgini F
- Subjects
- Animals, Cytoplasmic Granules drug effects, HEK293 Cells, Humans, Mice, N-Methylaspartate toxicity, Nerve Degeneration metabolism, Neurons drug effects, Neurons metabolism, Osmotic Pressure, Oxidative Stress drug effects, Protein Binding, Rats, Cytoplasmic Granules metabolism, Nerve Degeneration pathology, Parkinson Disease metabolism, Parkinson Disease pathology, Protein Deglycase DJ-1 metabolism, Ribonucleoproteins metabolism, Stress, Physiological drug effects
- Abstract
Mutations in the gene encoding DJ-1 are associated with autosomal recessive forms of Parkinson's disease (PD). DJ-1 plays a role in protection from oxidative stress, but how it functions as an "upstream" oxidative stress sensor and whether this relates to PD is still unclear. Intriguingly, DJ-1 may act as an RNA binding protein associating with specific mRNA transcripts in the human brain. Moreover, we previously reported that the yeast DJ-1 homolog Hsp31 localizes to stress granules (SGs) after glucose starvation, suggesting a role for DJ-1 in RNA dynamics. Here, we report that DJ-1 interacts with several SG components in mammalian cells and localizes to SGs, as well as P-bodies, upon induction of either osmotic or oxidative stress. By purifying the mRNA associated with DJ-1 in mammalian cells, we detected several transcripts and found that subpopulations of these localize to SGs after stress, suggesting that DJ-1 may target specific mRNAs to mRNP granules. Notably, we find that DJ-1 associates with SGs arising from N-methyl-D-aspartate (NMDA) excitotoxicity in primary neurons and parkinsonism-inducing toxins in dopaminergic cell cultures. Thus, our results indicate that DJ-1 is associated with cytoplasmic RNA granules arising during stress and neurodegeneration, providing a possible link between DJ-1 and RNA dynamics which may be relevant for PD pathogenesis.
- Published
- 2019
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.