33 results on '"Adriano Sebollela"'
Search Results
2. Neural Infection by Oropouche Virus in Adult Human Brain Slices Induces an Inflammatory and Toxic Response
- Author
-
Glaucia M. Almeida, Juliano P. Souza, Niele D. Mendes, Marjorie C. Pontelli, Nathalia R. Pinheiro, Giovanna O. Nogueira, Ricardo S. Cardoso, Isadora M. Paiva, Gustavo D. Ferrari, Flávio P. Veras, Fernando Q. Cunha, Jose A. C. Horta-Junior, Luciane C. Alberici, Thiago M. Cunha, Guilherme G. Podolsky-Gondim, Luciano Neder, Eurico Arruda, and Adriano Sebollela
- Subjects
arboviruses ,viral encephalitis ,histocultures ,neuroinflammation ,neuroinfection ,neurotropic virus ,Neurosciences. Biological psychiatry. Neuropsychiatry ,RC321-571 - Abstract
Oropouche virus (OROV) is an emerging arbovirus in South and Central Americas with high spreading potential. OROV infection has been associated with neurological complications and OROV genomic RNA has been detected in cerebrospinal fluid from patients, suggesting its neuroinvasive potential. Motivated by these findings, neurotropism and neuropathogenesis of OROV have been investigated in vivo in murine models, which do not fully recapitulate the complexity of the human brain. Here we have used slice cultures from adult human brains to investigate whether OROV is capable of infecting mature human neural cells in a context of preserved neural connections and brain cytoarchitecture. Our results demonstrate that human neural cells can be infected ex vivo by OROV and support the production of infectious viral particles. Moreover, OROV infection led to the release of the pro-inflammatory cytokine tumor necrosis factor-alpha (TNF-α) and diminished cell viability 48 h post-infection, indicating that OROV triggers an inflammatory response and tissue damage. Although OROV-positive neurons were observed, microglia were the most abundant central nervous system (CNS) cell type infected by OROV, suggesting that they play an important role in the response to CNS infection by OROV in the adult human brain. Importantly, we found no OROV-infected astrocytes. To the best of our knowledge, this is the first direct demonstration of OROV infection in human brain cells. Combined with previous data from murine models and case reports of OROV genome detection in cerebrospinal fluid from patients, our data shed light on OROV neuropathogenesis and help raising awareness about acute and possibly chronic consequences of OROV infection in the human brain.
- Published
- 2021
- Full Text
- View/download PDF
3. Morphological, cellular, and molecular basis of brain infection in COVID-19 patients
- Author
-
Fernanda Crunfli, Victor C. Carregari, Flavio P. Veras, Lucas S. Silva, Mateus Henrique Nogueira, André Saraiva Leão Marcelo Antunes, Pedro Henrique Vendramini, Aline Gazzola Fragnani Valença, Caroline Brandão-Teles, Giuliana da Silva Zuccoli, Guilherme Reis-de-Oliveira, Lícia C. Silva-Costa, Verônica Monteiro Saia-Cereda, Bradley J. Smith, Ana Campos Codo, Gabriela F de Souza, Stéfanie P. Muraro, Pierina Lorencini Parise, Daniel A. Toledo-Teixeira, Ícaro Maia Santos de Castro, Bruno Marcel Melo, Glaucia M. Almeida, Egidi Mayara Silva Firmino, Isadora Marques Paiva, Bruna Manuella Souza Silva, Rafaela Mano Guimarães, Niele D. Mendes, Raíssa L. Ludwig, Gabriel P. Ruiz, Thiago L. Knittel, Gustavo G. Davanzo, Jaqueline Aline Gerhardt, Patrícia Brito Rodrigues, Julia Forato, Mariene Ribeiro Amorim, Natália S. Brunetti, Matheus Cavalheiro Martini, Maíra Nilson Benatti, Sabrina S. Batah, Li Siyuan, Rafael B. João, Ítalo K. Aventurato, Mariana Rabelo de Brito, Maria J. Mendes, Beatriz A. da Costa, Marina K. M. Alvim, José Roberto da Silva Júnior, Lívia L. Damião, Iêda Maria P. de Sousa, Elessandra D. da Rocha, Solange M. Gonçalves, Luiz H. Lopes da Silva, Vanessa Bettini, Brunno M. Campos, Guilherme Ludwig, Lucas Alves Tavares, Marjorie Cornejo Pontelli, Rosa Maria Mendes Viana, Ronaldo B. Martins, Andre Schwambach Vieira, José Carlos Alves-Filho, Eurico Arruda, Guilherme Gozzoli Podolsky-Gondim, Marcelo Volpon Santos, Luciano Neder, André Damasio, Stevens Rehen, Marco Aurélio Ramirez Vinolo, Carolina Demarchi Munhoz, Paulo Louzada-Junior, Renê Donizeti Oliveira, Fernando Q. Cunha, Helder I. Nakaya, Thais Mauad, Amaro Nunes Duarte-Neto, Luiz Fernando Ferraz da Silva, Marisa Dolhnikoff, Paulo Hilario Nascimento Saldiva, Alessandro S. Farias, Fernando Cendes, Pedro Manoel M. Moraes-Vieira, Alexandre T. Fabro, Adriano Sebollela, José L. Proença-Modena, Clarissa L. Yasuda, Marcelo A. Mori, Thiago M. Cunha, and Daniel Martins-de-Souza
- Subjects
Post-Acute COVID-19 Syndrome ,Multidisciplinary ,SARS-CoV-2 ,Astrocytes ,Central Nervous System Viral Diseases ,Brain ,COVID-19 ,Humans - Abstract
Although increasing evidence confirms neuropsychiatric manifestations associated mainly with severe COVID-19 infection, long-term neuropsychiatric dysfunction (recently characterized as part of “long COVID-19” syndrome) has been frequently observed after mild infection. We show the spectrum of cerebral impact of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection, ranging from long-term alterations in mildly infected individuals (orbitofrontal cortical atrophy, neurocognitive impairment, excessive fatigue and anxiety symptoms) to severe acute damage confirmed in brain tissue samples extracted from the orbitofrontal region (via endonasal transethmoidal access) from individuals who died of COVID-19. In an independent cohort of 26 individuals who died of COVID-19, we used histopathological signs of brain damage as a guide for possible SARS-CoV-2 brain infection and found that among the 5 individuals who exhibited those signs, all of them had genetic material of the virus in the brain. Brain tissue samples from these five patients also exhibited foci of SARS-CoV-2 infection and replication, particularly in astrocytes. Supporting the hypothesis of astrocyte infection, neural stem cell–derived human astrocytes in vitro are susceptible to SARS-CoV-2 infection through a noncanonical mechanism that involves spike–NRP1 interaction. SARS-CoV-2–infected astrocytes manifested changes in energy metabolism and in key proteins and metabolites used to fuel neurons, as well as in the biogenesis of neurotransmitters. Moreover, human astrocyte infection elicits a secretory phenotype that reduces neuronal viability. Our data support the model in which SARS-CoV-2 reaches the brain, infects astrocytes, and consequently, leads to neuronal death or dysfunction. These deregulated processes could contribute to the structural and functional alterations seen in the brains of COVID-19 patients.
- Published
- 2022
- Full Text
- View/download PDF
4. AAV-mediated neuronal expression of a scFv antibody selective for Aβ oligomers protects synapses and rescues memory in Alzheimer models
- Author
-
Maria Clara Selles, Juliana T.S. Fortuna, Magali C. Cercato, Luis Eduardo Santos, Luciana Domett, Andre L.B. Bitencourt, Mariane Favero Carraro, Amanda S. Souza, Helena Janickova, Jorge M. de Souza, Soniza Alves-Leon, Vania F. Prado, Marco A. M. Prado, Alberto L. Epstein, Anna Salvetti, Ottavio Arancio, William L. Klein, Adriano Sebollela, Fernanda G. De Felice, Diana A. Jerusalinsky, and Sergio T. Ferreira
- Abstract
Brain accumulation of soluble oligomers of the amyloid-β peptide (AβOs) has been implicated in synapse failure and memory impairment in Alzheimer’s disease. Here, we show that treatment with NUsc1, a single-chain variable fragment antibody (scFv) that selectively targets AβOs, prevents the inhibition of long-term potentiation in hippocampal slices and memory impairment induced by AβOs in mice. As a therapeutic approach for intracerebral antibody delivery, we developed an adeno-associated virus vector to drive neuronal expression of NUsc1 (AAV-NUsc1) within the brain. Transduction by AAV-NUsc1 induced NUsc1 expression and secretion in adult human brain slices, and inhibited AβO binding to neurons and AβO-induced loss of dendritic spine loss in primary rat hippocampal cultures. Treatment of mice with AAV-NUsc1 prevented memory impairment induced by AβOs and, importantly, reversed memory deficits in aged APPswe/PS1ΔE9 Alzheimer’s disease model mice. These results support the feasibility of gene-mediated immunotherapy using single-chain antibodies as a potential therapeutic approach in Alzheimer’s disease.
- Published
- 2022
- Full Text
- View/download PDF
5. Simvastatin Prevents Long-Term Cognitive Deficits in Sepsis Survivor Rats by Reducing Neuroinflammation and Neurodegeneration
- Author
-
Carlos Henrique Rocha Catalão, Anderson de Oliveira Souza, Evelin Capellari Cárnio, Maria José Alves da Rocha, Adriano Sebollela, Luciane C. Alberici, Nilton Nascimento Santos-Junior, and Luís Henrique Angenendt da Costa
- Subjects
Male ,0301 basic medicine ,Simvastatin ,Hippocampus ,Pharmacology ,ASTRÓCITOS ,Toxicology ,Neuroprotection ,Sepsis ,03 medical and health sciences ,0302 clinical medicine ,medicine ,Animals ,Cognitive Dysfunction ,Rats, Wistar ,Prefrontal cortex ,Neuroinflammation ,Cognitive deficit ,Inflammation ,business.industry ,General Neuroscience ,Brain ,Neurodegenerative Diseases ,medicine.disease ,Rats ,030104 developmental biology ,Gliosis ,Hydroxymethylglutaryl-CoA Reductase Inhibitors ,medicine.symptom ,business ,030217 neurology & neurosurgery ,medicine.drug - Abstract
Sepsis-associated encephalopathy causes brain dysfunction that can result in cognitive impairments in sepsis survivor patients. In previous work, we showed that simvastatin attenuated oxidative stress in brain structures related to memory in septic rats. However, there is still a need to evaluate the long-term impact of simvastatin administration on brain neurodegenerative processes and cognitive damage in sepsis survivors. Here, we investigated the possible neuroprotective role of simvastatin in neuroinflammation, and neurodegeneration conditions of brain structures related to memory in rats at 10 days after sepsis survival. Male Wistar rats (250–300 g) were submitted to cecal ligation and puncture (CLP, n = 42) or remained as non-manipulated (naive, n = 30). Both groups were treated (before and after the surgery) by gavage with simvastatin (20 mg/kg) or an equivalent volume of saline and observed for 10 days. Simvastatin-treated rats that survived to sepsis showed a reduction in the levels of nitrate, IL1-β, and IL-6 and an increase in Bcl-2 protein expression in the prefrontal cortex and hippocampus, and synaptophysin only in the hippocampus. Immunofluorescence revealed a reduction of glial activation, neurodegeneration, apoptosis, and amyloid aggregates confirmed by quantification of GFAP, Iba-1, phospho Ser396-tau, total tau, cleaved caspase-3, and thioflavin-S in the prefrontal cortex and hippocampus. In addition, treated animals presented better performance in tasks involving habituation memory, discriminative, and aversive memory. These results suggest that statins exert a neuroprotective role by upregulation of the Bcl-2 and gliosis reduction, which may prevent the cognitive deficit observed in sepsis survivor animals.
- Published
- 2020
- Full Text
- View/download PDF
6. A genetic model of epilepsy with a partial Alzheimer’s disease-like phenotype and central insulin resistance
- Author
-
Suélen Santos Alves, Rui Milton Patrício da Silva Junior, Polianna Delfino-Pereira, Marilia Gabriella Alves Goulart Pereira, Israel Vasconcelos, Hanna Schwaemmle, Rodrigo Focosi Mazzei, Maiko Luiz Carlos, Enilza Maria Espreafico, Antônio Claudio Tedesco, Adriano Sebollela, Sebastião Sousa Almeida, José Antônio Cortes de Oliveira, and Norberto Garcia-Cairasco
- Subjects
Amyloid beta-Peptides ,Epilepsy ,Models, Genetic ,Neuroscience (miscellaneous) ,tau Proteins ,MEMÓRIA ,Rats ,Cellular and Molecular Neuroscience ,Phenotype ,Neurology ,Alzheimer Disease ,Animals ,Humans ,Insulin ,Insulin Resistance ,Rats, Wistar ,Maze Learning ,Aged - Abstract
Studies have suggested an important connection between epilepsy and Alzheimer's disease (AD), mostly due to the high number of patients diagnosed with AD who develop epileptic seizures later on. However, this link is not well understood. Previous studies from our group have identified memory impairment and metabolic abnormalities in the Wistar audiogenic rat (WAR) strain, a genetic model of epilepsy. Our goal was to investigate AD behavioral and molecular alterations, including brain insulin resistance, in naïve (seizure-free) animals of the WAR strain. We used the Morris water maze (MWM) test to evaluate spatial learning and memory performance and hippocampal tissue to verify possible molecular and immunohistochemical alterations. WARs presented worse performance in the MWM test (p 0.0001), higher levels of hyperphosphorylated tau (S396) (p 0.0001) and phosphorylated glycogen synthase kinase 3 (S21/9) (p 0.05), and lower insulin receptor levels (p 0.05). Conversely, WARs and Wistar controls present progressive increase in amyloid fibrils (p 0.0001) and low levels of soluble amyloid-β. Interestingly, the detected alterations were age-dependent, reaching larger differences in aged than in young adult animals. In summary, the present study provides evidence of a partial AD-like phenotype, including altered regulation of insulin signaling, in a genetic model of epilepsy. Together, these data contribute to the understanding of the connection between epilepsy and AD as comorbidities. Moreover, since both tau hyperphosphorylation and altered insulin signaling have already been reported in epilepsy and AD, these two events should be considered as important components in the interconnection between epilepsy and AD pathogenesis and, therefore, potential therapeutic targets in this field.
- Published
- 2022
7. Modeling the Human Brain With
- Author
-
Giovanna O, Nogueira, Patricia P, Garcez, Cedric, Bardy, Mark O, Cunningham, and Adriano, Sebollela
- Published
- 2021
8. A phylogenetically conserved hnRNP type A/B protein from squid brain
- Author
-
Diego Torrecillas Paula Lico, Roy E. Larson, Adriano Sebollela, Renata Rocha de Oliveira, Gabriel Sarti Lopes, Maria Luísa Paço-Larson, and Rafael Silva-Rocha
- Subjects
0301 basic medicine ,Cytoplasm ,Heterogeneous nuclear ribonucleoprotein ,Heterogeneous-Nuclear Ribonucleoproteins ,03 medical and health sciences ,0302 clinical medicine ,Stress granule ,Animals ,Humans ,NEURÔNIOS ,Peptide sequence ,Ribonucleoprotein ,Cell Nucleus ,Messenger RNA ,Chemistry ,General Neuroscience ,Decapodiformes ,Brain ,RNA-Binding Proteins ,Subcellular localization ,Protein tertiary structure ,Cell biology ,030104 developmental biology ,Ribonucleoproteins ,Dimerization ,030217 neurology & neurosurgery ,Nuclear localization sequence - Abstract
Eukaryotic mRNA precursors are co-transcriptionally assembled into ribonucleoprotein complexes. Heterogeneous nuclear ribonucleoprotein (hnRNP) complexes are involved in mRNA translocation, stability, subcellular localization and regulation of mRNA translation. About 20 major classes of hnRNPs have been identified in mammals. In a previous work, we characterized a novel, strongly-basic, RNA-binding protein (p65) in presynaptic terminals of squid neurons presenting homology with human hnRNPA/B type proteins, likely involved in local mRNA processing. We have identified and sequenced two hnRNPA/B-like proteins associated with tissue purified squid p65: Protein 1 (36.3 kDa, IP 7.1) and Protein 2 (37.6 kDa, IP 8.9). In the present work we generated an in silico, tridimensional, structural model of squid hnRNPA/B-like Protein 2, which showed highly conserved secondary and tertiary structure of RNA recognition motifs with human hnRNPA1 protein, as well as illustrated the potential for squid Protein 2 stable homodimerization. This was supported by biophysical measurements of bacterially expressed, recombinant protein. In addition, we induced expression of squid hnRNPA/B-like Protein 2 in human neuroblastoma cells (SH-SY5Y) and observed an exclusively nuclear localization, which depended on an intact C-terminal amino acid sequence and which relocated to cytoplasm particles containing PABP when the cells were challenged with sorbitol, suggesting an involvement with stress granule function.
- Published
- 2019
- Full Text
- View/download PDF
9. A freeze-and-thaw-induced fragment of the microtubule-associated protein tau in rat brain extracts: implications for the biochemical assessment of neurotoxicity
- Author
-
Adriano Sebollela, Gustavo Duarte Ferrari, Hanna K. Schwaemmle, Israel C. Vasconcelos, Luciane C. Alberici, N. Garcia-Cairasco, Raquel M. Campos, Ana Paula Masson, and Vitor M. Faça
- Subjects
0301 basic medicine ,medicine.medical_treatment ,Phosphatase ,Tau protein ,Biophysics ,tau Proteins ,Biochemistry ,Molecular Bases of Health & Disease ,tau protein ,03 medical and health sciences ,0302 clinical medicine ,Western blot ,Biochemical Techniques & Resources ,Alzheimer Disease ,medicine ,Animals ,Humans ,Rats, Wistar ,Molecular Biology ,Research Articles ,Cryopreservation ,METABOLISMO DE PROTEÍNA ,Protease ,disease models ,western blot ,medicine.diagnostic_test ,biology ,Chemistry ,tauopathy ,Neurotoxicity ,neurodegeneration ,Brain ,Cell Biology ,Alzheimer's disease ,medicine.disease ,Molecular biology ,Immunohistochemistry ,Rats ,Blot ,030104 developmental biology ,Radioimmunoprecipitation assay buffer ,Proteolysis ,biology.protein ,Tauopathy ,030217 neurology & neurosurgery - Abstract
Tau is a microtubule-associated protein (MAP) responsible for controlling the stabilization of microtubules in neurons. Tau function is regulated by phosphorylation. However, in some neurological diseases Tau becomes aberrantly hyperphosphorylated, which contributes to the pathogenesis of neurological diseases, known as tauopathies. Western blotting (WB) has been widely employed to determine Tau levels in neurological disease models. However, Tau quantification by WB should be interpreted with care, as this approach has been recognized as prone to produce artifactual results if not properly performed. In the present study, our goal was to evaluate the influence of a freeze-and-thaw cycle, a common procedure preceding WB, to the integrity of Tau in brain homogenates from rats, 3xTg-AD mice and human samples. Homogenates were prepared in ice-cold RIPA buffer supplemented with protease/phosphatase inhibitors. Immediately after centrifugation, an aliquot of the extracts was analyzed via WB to quantify total and phosphorylated Tau levels. The remaining aliquots of the same extracts were stored for at least 2 weeks at either −20 or −80°C and then subjected to WB. Extracts from rodent brains submitted to freeze-and-thaw presented a ∼25 kDa fragment immunoreactive to anti-Tau antibodies. An in-gel digestion followed by mass spectrometry (MS) analysis in excised bands revealed this ∼25 kDa species corresponds to a Tau fragment. Freeze-and-thaw-induced Tau proteolysis was detected even when extracts were stored at −80°C. This phenomenon was not observed in human samples at any storage condition tested. Based on these findings, we strongly recommend the use of fresh extracts of brain samples in molecular analysis of Tau levels in rodents.
- Published
- 2020
10. SARS-CoV-2 infects brain astrocytes of COVID-19 patients and impairs neuronal viability
- Author
-
Bruna Manuella Souza Silva, Jaqueline Aline Gerhardt, Iêda Maria Pereira de Sousa, Aline Gazzola Fragnani Valenca, Alessandro S. Farias, M. C. Martini, Maria Ercilia de Paula Castilho Stefano, Vanessa Bettini, Eurico de Arruda Neto, Marjorie Cornejo Pontelli, Paulo Louzada-Junior, Amaro Nunes Duarte-Neto, Guilherme Podolski-Gondim, Bradley J. Smith, Clarissa L. Yasuda, Marina K. M. Alvim, Solange Maria Gonçalves, Marcelo A. Mori, Thais Mauad, Raissa G. Ludwig, Mateus Henrique Nogueira, Elessandra Dias da Rocha, Natália Brunetti Silva, Isadora Marques Paiva, Daniel A. Toledo-Teixeira, André Schwambach Vieira, Ana Campos Codo, Patrícia Brito Rodrigues, Lucas Alves Tavares, José Roberto da Silva Junior, Adriano Sebollela, Fernando Q. Cunha, Niele D. Mendes, Guilherme Ludwig, Gustavo Gastão Davanzo, Stevens K. Rehen, André Saraiva Leão Marcelo Antunes, Glaucia M. Almeida, Verônica M. Saia-Cereda, Daniel Martins-de-Souza, Julia Forato, Lucas Scardua Silva, Egidi Mayara Silva Firmino, Stéfanie Primon Muraro, Bruno Marcel Silva de Melo, Rosa Maria Mendes Viana, Ronaldo B. Martins, Marco Aurélio Ramirez Vinolo, Pedro M. Moraes-Vieira, Ícaro Maia Santos de Castro, Carolina Brandao-Teles, Helder I. Nakaya, Guilherme Reis-de-Oliveira, Lívia Liviane Damião, ítalo Karmann Aventurato, Li Siyuan, Fernando Cendes, Marcelo Volpon Santos, Pierina Lorencini Parise, Carolina Demarchi Munhoz, Pedro Henrique Vendramini, Mariana Rabelo de Brito, Sabrina Setembre Batah, José C. Alves-Filho, Fernanda Crunfli, Gabriel Palermo Ruiz, Victor Corasolla Carregari, Giuliana S. Zuccoli, Flávio P. Veras, Paulo Hilário Nascimento Saldiva, Licia C. Silva-Costa, Maira N. Benatti, Luiz Henrique Lopes da Silva, Gabriela Fabiano de Souza, Rafaela M. Guimarães, Brunno Machado de Campos, Rafael Batista João, Thiago M. Cunha, Luiz Fernando Ferraz da Silva, Thiago L. Knittel, Luciano Neder, José Luiz Proença Modena, André Damasio, Marisa Dolhnikoff, Renê Donizeti Ribeiro de Oliveira, Alexandre Todorovic Fabro, and Mariene R. Amorim
- Subjects
Pathology ,medicine.medical_specialty ,Coronavirus disease 2019 (COVID-19) ,business.industry ,Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) ,Energy metabolism ,Brain damage ,Phenotype ,medicine ,Respiratory system ,medicine.symptom ,business ,Cognitive impairment ,Biogenesis - Abstract
COVID-19 patients may exhibit neuropsychiatric and neurological symptoms. We found that anxiety and cognitive impairment are manifested by 28-56% of SARS-CoV-2-infected individuals with mild respiratory symptoms and are associated with altered cerebral cortical thickness. Using an independent cohort, we found histopathological signs of brain damage in 25% of individuals who died of COVID-19. All of the affected brain tissues exhibited foci of SARS-CoV-2 infection and replication, particularly in astrocytes. Infection of neural stem cell-derived astrocytes changed energy metabolism, altered key proteins and metabolites used to fuel neurons and for biogenesis of neurotransmitters, and elicited a secretory phenotype that reduces neuronal viability. Our data support the model where SARS-CoV-2 reaches the brain, infects astrocytes and triggers neuropathological changes that contribute to the structural and functional alterations in the brain of COVID-19 patients.
- Published
- 2020
- Full Text
- View/download PDF
11. Short-Term Free-Floating Slice Cultures from the Adult Human Brain
- Author
-
Norberto Garcia-Cairasco, Luciano Neder, Niele D. Mendes, Artur Fernandes, João Alberto Assirati Junior, Adriano Sebollela, Giovanna Orlovski Nogueira, José de Anchieta C. Horta-Júnior, Carla Machado, Glaucia M. Almeida, Universidade de São Paulo (USP), and Universidade Estadual Paulista (Unesp)
- Subjects
0301 basic medicine ,Adult ,General Chemical Engineering ,Central nervous system ,Stimulus (physiology) ,Biology ,General Biochemistry, Genetics and Molecular Biology ,Temporal lobe ,histoculture ,03 medical and health sciences ,0302 clinical medicine ,Organ Culture Techniques ,parasitic diseases ,medicine ,neocortex ,Animals ,Humans ,Neural cell ,Neurons ,Neocortex ,Cell Death ,General Immunology and Microbiology ,Nervous tissue ,General Neuroscience ,Neurodegeneration ,neurodegeneration ,Brain ,Human brain ,Hydrogen Peroxide ,medicine.disease ,Alzheimer's ,Issue 153 ,030104 developmental biology ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,ex vivo ,epilepsy ,Neuroscience ,DOENÇA DE ALZHEIMER - Abstract
Made available in DSpace on 2020-12-10T20:13:04Z (GMT). No. of bitstreams: 0 Previous issue date: 2019-11-01 Fundação de Amparo à Pesquisa do Estado de São Paulo (FAPESP) Coordenação de Aperfeiçoamento de Pessoal de Nível Superior (CAPES) FAEPA Conselho Nacional de Desenvolvimento Científico e Tecnológico (CNPq) Organotypic, or slice cultures, have been widely employed to model aspects of the central nervous system functioning in vitro. Despite the potential of slice cultures in neuroscience, studies using adult nervous tissue to prepare such cultures are still scarce, particularly those from human subjects. The use of adult human tissue to prepare slice cultures is particularly attractive to enhance the understanding of human neuropathologies, as they hold unique properties typical of the mature human brain lacking in slices produced from rodent (usually neonatal) nervous tissue. This protocol describes how to use brain tissue collected from living human donors submitted to resective brain surgery to prepare short-term, free-floating slice cultures. Procedures to maintain and perform biochemical and cell biology assays using these cultures are also presented. Representative results demonstrate that the typical human cortical lamination is preserved in slices after 4 days in vitro (DIV4), with expected presence of the main neural cell types. Moreover, slices at DIV4 undergo robust cell death when challenged with a toxic stimulus (H2O2), indicating the potential of this model to serve as a platform in cell death assays. This method, a simpler and cost-effective alternative to the widely used protocol using membrane inserts, is mainly recommended for running short-term assays aimed to unravel mechanisms of neurodegeneration behind age-associated brain diseases. Finally, although the protocol is devoted to using cortical tissue collected from patients submitted to surgical treatment of pharmacoresistant temporal lobe epilepsy, it is argued that tissue collected from other brain regions/conditions should also be considered as sources to produce similar free-floating slice cultures. Univ Sao Paulo, Ribeirao Preto Med Sch, Dept Biochem & Immunol, Sao Paulo, SP, Brazil Univ Sao Paulo, Ribeirao Preto Med Sch, Dept Physiol, Sao Paulo, SP, Brazil Univ Sao Paulo, Ribeirao Preto Med Sch, Dept Pathol & Forens Med, Sao Paulo, SP, Brazil Sao Paulo State Univ, Inst Biosci, Dept Anat, Sao Paulo, SP, Brazil Univ Sao Paulo, Ribeirao Preto Med Sch, Clin Hosp, Sao Paulo, SP, Brazil Sao Paulo State Univ, Inst Biosci, Dept Anat, Sao Paulo, SP, Brazil FAPESP: MS 2018/06614-4 FAPESP: 25681-3/2017
- Published
- 2019
- Full Text
- View/download PDF
12. Antibody Fragments as Tools for Elucidating Structure-Toxicity Relationships and for Diagnostic/Therapeutic Targeting of Neurotoxic Amyloid Oligomers
- Author
-
Erika N. Cline, William L. Klein, Raquel M. Campos, Adriano Sebollela, and André L B Bitencourt
- Subjects
Central Nervous System ,Amyloid ,Review ,Protein Aggregation, Pathological ,Oligomer ,oligomer ,Catalysis ,Antibody fragments ,lcsh:Chemistry ,Inorganic Chemistry ,Immunoglobulin Fab Fragments ,Structure-Activity Relationship ,chemistry.chemical_compound ,neurotoxicity ,PATOLOGIA ,medicine ,Animals ,Humans ,Physical and Theoretical Chemistry ,Immunoglobulin Fragments ,lcsh:QH301-705.5 ,Molecular Biology ,Spectroscopy ,antibody fragments ,biology ,single chain ,Organic Chemistry ,Neurotoxicity ,amyloid ,Amyloidosis ,General Medicine ,Single-Domain Antibodies ,medicine.disease ,Ligand (biochemistry) ,Peptide Fragments ,In vitro ,Computer Science Applications ,NUsc1 ,lcsh:Biology (General) ,lcsh:QD1-999 ,chemistry ,Biochemistry ,Toxicity ,biology.protein ,Neurotoxicity Syndromes ,Antibody - Abstract
The accumulation of amyloid protein aggregates in tissues is the basis for the onset of diseases known as amyloidoses. Intriguingly, many amyloidoses impact the central nervous system (CNS) and usually are devastating diseases. It is increasingly apparent that neurotoxic soluble oligomers formed by amyloidogenic proteins are the primary molecular drivers of these diseases, making them lucrative diagnostic and therapeutic targets. One promising diagnostic/therapeutic strategy has been the development of antibody fragments against amyloid oligomers. Antibody fragments, such as fragment antigen-binding (Fab), scFv (single chain variable fragments), and VHH (heavy chain variable domain or single-domain antibodies) are an alternative to full-length IgGs as diagnostics and therapeutics for a variety of diseases, mainly because of their increased tissue penetration (lower MW compared to IgG), decreased inflammatory potential (lack of Fc domain), and facile production (low structural complexity). Furthermore, through the use of in vitro-based ligand selection, it has been possible to identify antibody fragments presenting marked conformational selectivity. In this review, we summarize significant reports on antibody fragments selective for oligomers associated with prevalent CNS amyloidoses. We discuss promising results obtained using antibody fragments as both diagnostic and therapeutic agents against these diseases. In addition, the use of antibody fragments, particularly scFv and VHH, in the isolation of unique oligomeric assemblies is discussed as a strategy to unravel conformational moieties responsible for neurotoxicity. We envision that advances in this field may lead to the development of novel oligomer-selective antibody fragments with superior selectivity and, hopefully, good clinical outcomes.
- Published
- 2020
- Full Text
- View/download PDF
13. A freeze-and-thaw induced-fragment of the microtubule-associated protein Tau in rat brain extracts: implications for the biochemical assessment of neurotoxicity
- Author
-
N. Garcia-Cairasco, Luciane C. Alberici, Ana Paula Masson, Adriano Sebollela, Gustavo Duarte Ferrari, Raquel M. Campos, Israel C. Vasconcelos, Hanna K. Schwaemmle, and Vitor M. Faça
- Subjects
Protease ,medicine.diagnostic_test ,Chemistry ,Proteolysis ,medicine.medical_treatment ,Phosphatase ,Neurotoxicity ,medicine.disease ,Molecular biology ,Blot ,Radioimmunoprecipitation assay buffer ,medicine ,Phosphorylation ,Centrifugation - Abstract
Tau is a microtubule-associated protein responsible for controlling the stabilization of microtubules in neurons. Tau function is regulated by phosphorylation. However, in some neurological diseases Tau becomes aberrantly hyperphosphorylated, which contributes to the pathogenesis of neurological diseases, known as tauopathies. Western blotting (WB) has been widely employed to determine Tau levels in neurological disease models. However, Tau quantification by WB should be interpreted with care, as this approach has been recognized as prone to produce artifactual results if not properly performed. In this study, our goal was to evaluate the influence of a freeze-and-thaw cycle, a common procedure preceding WB, to the integrity of Tau in brain homogenates from rats, 3xTg-AD mice and human samples. Homogenates were prepared in ice-cold RIPA buffer supplemented with protease/phosphatase inhibitors. Immediately after centrifugation, an aliquot of the extracts was analyzed via WB to quantify total and phosphorylated Tau levels. The remaining aliquots were stored for at least 2 weeks at either −20°C or −80°C and then subjected to WB. Extracts from rodent brains submitted to freeze-and-thaw presented a ~25 kDa fragment immunoreactive to anti-Tau antibodies. An in-gel digestion followed by mass spectrometry analysis in excised bands revealed this ~25 kDa species corresponds to a Tau fragment. Freeze-and-thaw-induced Tau proteolysis was detected even when extracts were stored at −80°C. This phenomenon was not observed in human samples at any storage condition tested. Based on these findings, we strongly recommend the use of fresh extracts of brain samples in molecular analysis of Tau levels in rodents.
- Published
- 2018
- Full Text
- View/download PDF
14. Free-floating adult human brain-derived slice cultures as a model to study the neuronal impact of Alzheimer’s disease-associated Aβ oligomers
- Author
-
Adriano Sebollela, N.M. Lyra e Silva, Jorge Marcondes, João Alberto Assirati, Luciano Neder, Glaucia M. Almeida, Norberto Garcia-Cairasco, William L. Klein, Sergio T. Ferreira, Caio M. Matias, Paulo Roberto Louzada, Soniza Vieira Alves-Leon, José de Anchieta C. Horta-Júnior, Artur Fernandes, Carla Machado, Fernanda G. De Felice, Niele D. Mendes, Maria Clara Selles, Luis E. Santos, Universidade de São Paulo (USP), Universidade Federal do Rio de Janeiro (UFRJ), Universidade Estadual Paulista (Unesp), Queens Univ, Northwestern Univ, and Barretos Canc Hosp
- Subjects
0301 basic medicine ,Adult ,Male ,NEUROCIÊNCIAS ,tau Proteins ,Biology ,In Vitro Techniques ,Human brain ,Potassium Chloride ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Organ Culture Techniques ,parasitic diseases ,medicine ,Premovement neuronal activity ,Humans ,Viability assay ,Neurotransmitter ,Organotypic culture ,Cerebral Cortex ,Neurons ,Analysis of Variance ,Neurotransmitter Agents ,Amyloid beta-Peptides ,Epilepsy ,A beta oligomers ,General Neuroscience ,Neurotoxicity ,virus diseases ,Middle Aged ,Tissue slices ,Alzheimer's disease ,medicine.disease ,Cortex (botany) ,030104 developmental biology ,medicine.anatomical_structure ,chemistry ,Cytoarchitecture ,Epilepsy, Temporal Lobe ,Cell culture ,Phosphopyruvate Hydratase ,Female ,Neuroscience ,030217 neurology & neurosurgery ,geographic locations - Abstract
Made available in DSpace on 2018-11-26T22:40:48Z (GMT). No. of bitstreams: 0 Previous issue date: 2018-09-01 Fundação de Amparo à Pesquisa do Estado de São Paulo (FAPESP) FAEPA National Institute for Translational Neuroscience Fundação de Amparo à Pesquisa do Estado do Rio de Janeiro (FAPERJ) Conselho Nacional de Desenvolvimento Científico e Tecnológico (CNPq) Coordenação de Aperfeiçoamento de Pessoal de Nível Superior (CAPES) Background: Slice cultures have been prepared from several organs. With respect to the brain, advantages of slice cultures over dissociated cell cultures include maintenance of the cytoarchitecture and neuronal connectivity. Slice cultures from adult human brain have been reported and constitute a promising method to study neurological diseases. Despite this potential, few studies have characterized in detail cell survival and function along time in short-term, free-floating cultures. New Method: We used tissue from adult human brain cortex from patients undergoing temporal lobectomy to prepare 200 pm-thick slices. Along the period in culture, we evaluated neuronal survival, histological modifications, and neurotransmitter release. The toxicity of Alzheimer's-associated A beta oligomers (A beta Os) to cultured slices was also analyzed. Results: Neurons in human brain slices remain viable and neurochemically active for at least four days in vitro, which allowed detection of binding of A beta Os. We further found that slices exposed to A beta Os presented elevated levels of hyperphosphorylated Tau, a hallmark of Alzheimer's disease. Comparison with Existing Method(s): Although slice cultures from adult human brain have been previously prepared, this is the first report to analyze cell viability and neuronal activity in short-term free-floating cultures as a function of days in vitro. Conclusions: Once surgical tissue is available, the current protocol is easy to perform and produces functional slices from adult human brain. These slice cultures may represent a preferred model for translational studies of neurodegenerative disorders when long term culturing in not required, as in investigations on A beta O neurotoxicity. Univ Sao Paulo, Ribeirao Preto Med Sch, Dept Biochem & Immunol, Ribeirao Preto, SP, Brazil Univ Sao Paulo, Ribeirao Preto Med Sch, Dept Pathol & Forens Med, Ribeirao Preto, SP, Brazil Univ Sao Paulo, Ribeirao Preto Med Sch, Dept Physiol, Ribeirao Preto, SP, Brazil Univ Fed Rio de Janeiro, Inst Med Biochem, Rio De Janeiro, RJ, Brazil Sao Paulo State Univ, Inst Biosci, Dept Anat, Sao Paulo, Brazil Univ Fed Rio de Janeiro, Inst Biomed Sci, Rio De Janeiro, RJ, Brazil Queens Univ, Ctr Neurosci Studies, Dept Biomed & Mol Sci, Kingston, ON, Canada Univ Fed Rio de Janeiro, Hosp Univ Clementino Fraga Filho, Rio De Janeiro, RJ, Brazil Univ Sao Paulo, Ribeirao Preto Med Sch, Clin Hosp, Ribeirao Preto, SP, Brazil Northwestern Univ, Dept Neurobiol, Evanston, IL 60208 USA Univ Fed Rio de Janeiro, Inst Biophys Carlos Chagas Filho, Rio De Janeiro, RJ, Brazil Barretos Canc Hosp, Barretos, SP, Brazil Sao Paulo State Univ, Inst Biosci, Dept Anat, Sao Paulo, Brazil
- Published
- 2018
15. Neuronal expression of NUsc1, a single-chain variable fragment antibody against Ab oligomers, protects synapses and rescues memory in Alzheimer's disease models
- Author
-
Diana Jerusalinsky, Amanda Santos de Souza, Marco A. M. Prado, William L. Klein, Ottavio Arancio, Sergio T. Ferreira, Magalí Cecilia Cercato, Vania F. Prado, Juliana T.S. Fortuna, Adriano Sebollela, Fernanda G. De Felice, Maria Clara Selles, and André L B Bitencourt
- Subjects
Chemistry ,Single-Chain Variable Fragment Antibody ,General Neuroscience ,Disease ,Cell biology - Published
- 2019
- Full Text
- View/download PDF
16. Memantine Rescues Transient Cognitive Impairment Caused by High-Molecular-Weight A Oligomers But Not the Persistent Impairment Induced by Low-Molecular-Weight Oligomers
- Author
-
Jose Henrique Ledo, Carine V Costa, Julia R. Clarke, Helen M. Melo, Fernanda G. De Felice, Sergio T. Ferreira, Leonardo M. Saraiva, Adriano Sebollela, Axa P Mota-Sales, Felipe C. Ribeiro, William L. Klein, and Cláudia P. Figueiredo
- Subjects
Male ,medicine.medical_specialty ,Hippocampal formation ,medicine.disease_cause ,Oligomer ,Synapse ,Mice ,chemistry.chemical_compound ,Memantine ,Internal medicine ,medicine ,Animals ,Memory impairment ,Cells, Cultured ,Cognitive deficit ,Amyloid beta-Peptides ,biology ,Chemistry ,General Neuroscience ,food and beverages ,Articles ,Peptide Fragments ,Rats ,Molecular Weight ,Endocrinology ,Synaptophysin ,biology.protein ,medicine.symptom ,Cognition Disorders ,Neuroscience ,Oxidative stress ,medicine.drug - Abstract
Brain accumulation of soluble amyloid-β oligomers (AβOs) has been implicated in synapse failure and cognitive impairment in Alzheimer's disease (AD). However, whether and how oligomers of different sizes induce synapse dysfunction is a matter of controversy. Here, we report that low-molecular-weight (LMW) and high-molecular-weight (HMW) Aβ oligomers differentially impact synapses and memory. A single intracerebroventricular injection of LMW AβOs (10 pmol) induced rapid and persistent cognitive impairment in mice. On the other hand, memory deficit induced by HMW AβOs (10 pmol) was found to be reversible. While memory impairment in LMW oligomer-injected mice was associated with decreased hippocampal synaptophysin and GluN2B immunoreactivities, synaptic pathology was not detected in the hippocampi of HMW oligomer-injected mice. On the other hand, HMW oligomers, but not LMW oligomers, induced oxidative stress in hippocampal neurons. Memantine rescued both neuronal oxidative stress and the transient memory impairment caused by HMW oligomers, but did not prevent the persistent cognitive deficit induced by LMW oligomers. Results establish that different Aβ oligomer assemblies act in an orchestrated manner, inducing different pathologies and leading to synapse dysfunction. Furthermore, results suggest a mechanistic explanation for the limited efficacy of memantine in preventing memory loss in AD.
- Published
- 2013
- Full Text
- View/download PDF
17. A human scFv antibody that targets and neutralizes high molecular weight pathogenic amyloid-β oligomers
- Author
-
Lei A. Qin, Josette M. Kamel, Izolda A. Popova, Kirsten L. Viola, Kevin Luo, Anthea Weng, Milena A. Barcelos, Adrian M. Bebenek, William L. Klein, Erika N. Cline, Adriano Sebollela, Vanessa N. Bezerra, Pauline T. Velasco, Natalia M. Lyra e Silva, Nadia DiNunno, Jay Ahn, Pascale N. Lacor, Xiao-xia Sun, Jason Patel, Sergio T. Ferreira, and Nathalia R. Pinheiro
- Subjects
ANTICORPOS ,0301 basic medicine ,chemistry.chemical_classification ,biology ,Molecular mass ,Wild type ,Endogeny ,Peptide ,Hippocampal formation ,medicine.disease_cause ,Biochemistry ,Pathogenesis ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,030104 developmental biology ,0302 clinical medicine ,chemistry ,biology.protein ,medicine ,Antibody ,030217 neurology & neurosurgery ,Oxidative stress - Abstract
Brain accumulation of soluble oligomers of the amyloid-β peptide (AβOs) is increasingly considered a key early event in the pathogenesis of Alzheimer's disease (AD). A variety of AβO species have been identified, both in vitro and in vivo, ranging from dimers to 24mers and higher order oligomers. However, there is no consensus in the literature regarding which AβO species are most germane to AD pathogenesis. Antibodies capable of specifically recognizing defined subpopulations of AβOs would be a valuable asset in the identification, isolation, and characterization of AD-relevant AβO species. Here, we report the characterization of a human single chain antibody fragment (scFv) denoted NUsc1, one of a number of scFvs we have identified that stringently distinguish AβOs from both monomeric and fibrillar Aβ. NUsc1 readily detected AβOs previously bound to dendrites in cultured hippocampal neurons. In addition, NUsc1 blocked AβO binding and reduced AβO-induced neuronal oxidative stress and tau hyperphosphorylation in cultured neurons. NUsc1 further distinguished brain extracts from AD-transgenic mice from wild type (WT) mice, and detected endogenous AβOs in fixed AD brain tissue and AD brain extracts. Biochemical analyses indicated that NUsc1 targets a subpopulation of AβOs with apparent molecular mass greater than 50 kDa. Results indicate that NUsc1 targets a particular AβO species relevant to AD pathogenesis, and suggest that NUsc1 may constitute an effective tool for AD diagnostics and therapeutics.
- Published
- 2017
18. Expression Profile of Rat Hippocampal Neurons Treated with the Neuroprotective Compound 2,4-Dinitrophenol: Up-Regulation of cAMP Signaling Genes
- Author
-
Juliana Camacho-Pereira, Helena Brentani, Ana Paula Wasilewska-Sampaio, Camila T. Mendes, Adriano Sebollela, Sergio T. Ferreira, Emmanuel Dias-Neto, Léo Freitas-Correa, Fabio Passetti, Antonio Galina, Fernanda G. De Felice, and Fabio F. Oliveira
- Subjects
Neurite ,Cell Survival ,Cell Culture Techniques ,Down-Regulation ,chemical and pharmacologic phenomena ,Hippocampal formation ,Biology ,Toxicology ,Hippocampus ,Neuroprotection ,Adenosine Triphosphate ,Oxygen Consumption ,Downregulation and upregulation ,Gene expression ,Cyclic AMP ,Animals ,Gene ,Oligonucleotide Array Sequence Analysis ,Dose-Response Relationship, Drug ,Gene Expression Profiling ,General Neuroscience ,Rats ,Up-Regulation ,Adenosine Diphosphate ,Neuroprotective Agents ,Biochemistry ,Synaptic plasticity ,2,4-Dinitrophenol ,Reactive Oxygen Species ,Intracellular ,Signal Transduction - Abstract
2,4-Dinitrophenol (DNP) is classically known as a mitochondrial uncoupler and, at high concentrations, is toxic to a variety of cells. However, it has recently been shown that, at subtoxic concentrations, DNP protects neurons against a variety of insults and promotes neuronal differentiation and neuritogenesis. The molecular and cellular mechanisms underlying the beneficial neuroactive properties of DNP are still largely unknown. We have now used DNA microarray analysis to investigate changes in gene expression in rat hippocampal neurons in culture treated with low micromolar concentrations of DNP. Under conditions that did not affect neuronal viability, high-energy phosphate levels or mitochondrial oxygen consumption, DNP induced up-regulation of 275 genes and down-regulation of 231 genes. Significantly, several up-regulated genes were linked to intracellular cAMP signaling, known to be involved in neurite outgrowth, synaptic plasticity, and neuronal survival. Differential expression of specific genes was validated by quantitative RT-PCR using independent samples. Results shed light on molecular mechanisms underlying neuroprotection by DNP and point to possible targets for development of novel therapeutics for neurodegenerative disorders.
- Published
- 2009
- Full Text
- View/download PDF
19. Interrogating global gene expression in rat neuronal cultures using SAGE
- Author
-
Adriano Sebollela, Sergio T. Ferreira, and Emmanuel Dias-Neto
- Subjects
Combined use ,Gene Expression ,Biology ,Toxicology ,Neuroprotection ,Gene expression ,medicine ,Transcriptional regulation ,Animals ,Serial analysis of gene expression ,Molecular Biology ,Cells, Cultured ,Gene Library ,Neurons ,General Neuroscience ,SAGE ,fungi ,Neurotoxicity ,DNA ,medicine.disease ,Rats ,Neuroprotective Agents ,Molecular targets ,RNA ,Poly A ,Neuroscience - Abstract
The normal function of the mammalian brain is regulated by complex networks of interactions between cells and molecules, which are to a con|siderable extent dependent on mechanisms of transcriptional regulation. Disruption of such interactions by neurotoxic stimuli may lead to severe forms of dementia and to other neuropsychiatric disorders. Therefore, critical insight into mechanisms of neuronal dysfunction may be obtained by examining global patterns of gene expression in mammalian models of neurotoxicity. In this regard, the combined use of rat neuronal cultures and serial analysis of gene expression (SAGE) can be viewed as a general platform to enable the search for molecular targets involved in neurotoxic processes. Here, we discuss potential advantages of this approach, highlighting the need for generation of robust SAGE libraries from rat neuronal cultures. The availability and current limitations of bioinformatics tools for SAGE data derived from rat samples is also discussed.
- Published
- 2007
- Full Text
- View/download PDF
20. Soluble oligomers from a non-disease related protein mimic Aβ-induced tau hyperphosphorylation and neurodegeneration
- Author
-
Sergio T. Ferreira, Leonardo M. Saraiva, Jean-Christophe Houzel, Leticia Forny-Germano, Marcelo N. N. Vieira, Fernanda G. De Felice, Adriano Sebollela, and Ana Maria Blanco Martinez
- Subjects
chemistry.chemical_classification ,biology ,Amyloid ,Amyloidosis ,Neurodegeneration ,Tau protein ,Peptide ,Protein aggregation ,medicine.disease ,Biochemistry ,Cellular and Molecular Neuroscience ,Amyloid disease ,chemistry ,medicine ,biology.protein ,Biophysics ,Alzheimer's disease - Abstract
Protein aggregation and amyloid accumulation in different tissues are associated with cellular dysfunction and toxicity in important human pathologies, including Alzheimer's disease and various forms of systemic amyloidosis. Soluble oligomers formed at the early stages of protein aggregation have been increasingly recognized as the main toxic species in amyloid diseases. To gain insight into the mechanisms of toxicity instigated by soluble protein oligomers, we have investigated the aggregation of hen egg white lysozyme (HEWL), a normally harmless protein. HEWL initially aggregates into beta-sheet rich, roughly spherical oligomers which appear to convert with time into protofibrils and mature amyloid fibrils. HEWL oligomers are potently neurotoxic to rat cortical neurons in culture, while mature amyloid fibrils are little or non-toxic. Interestingly, when added to cortical neuronal cultures HEWL oligomers induce tau hyperphosphorylation at epitopes that are characteristically phosphorylated in neurons exposed to soluble oligomers of the amyloid-beta peptide. Furthermore, injection of HEWL oligomers in the cerebral cortices of adult rats induces extensive neurodegeneration in different brain areas. These results show that soluble oligomers from a non-disease related protein can mimic specific neuronal pathologies thought to be induced by soluble amyloid-beta peptide oligomers in Alzheimer's disease and support the notion that amyloid oligomers from different proteins may share common structural determinants that would explain their generic cytotoxicities.
- Published
- 2007
- Full Text
- View/download PDF
21. Heparin-binding Sites in Granulocyte-Macrophage Colony-stimulating Factor
- Author
-
Marcos Henrique Ferreira Sorgine, Carlos H.I. Ramos, Gabriel S. C.S. Limaverde, Thiago C. Cagliari, Sergio T. Ferreira, Adriano Sebollela, Tatiana Coelho-Sampaio, and Alex Chapeaurouge
- Subjects
Alanine ,Stereochemistry ,Wild type ,Cell Biology ,Heparan sulfate ,Heparin ,Biochemistry ,chemistry.chemical_compound ,Residue (chemistry) ,Sulfation ,chemistry ,medicine ,Binding site ,Molecular Biology ,Histidine ,medicine.drug - Abstract
The biological activity of granulocyte-macrophage colony-stimulating factor (GM-CSF) is modulated by the sulfated glycosaminoglycans (GAGs) heparan sulfate and heparin. However, the molecular mechanisms involved in such interactions are still not completely understood. We have proposed previously that helix C, one of the four α-helices of human GM-CSF (hGM-CSF), contains a GAG-binding site in which positively charged residues are spatially positioned for interaction with the sulfate moieties of the GAGs (Wettreich, A., Sebollela, A., Carvalho, M. A., Azevedo, S. P., Borojevic, R., Ferreira, S. T., and Coelho-Sampaio, T. (1999) J. Biol. Chem. 274, 31468-31475). Protonation of two histidine residues (His83 and His87) in helix C of hGM-CSF appears to act as a pH-dependent molecular switch to control the interaction with GAGs. Based on these findings, we have now generated a triple mutant form of murine GM-CSF (mGM-CSF) in which three noncharged residues in helix C of the murine factor (Tyr83, Gln85, and Tyr87) were replaced by the corresponding basic residues present in hGM-CSF (His83, Lys85, and His87). Binding assays on heparin-Sepharose showed that, at acidic pH, the triple mutant mGM-CSF binds to immobilized heparin with significantly higher affinity than wild type (WT) mGM-CSF and that neither protein binds to the column at neutral pH. The fact that even WT mGM-CSF binds to heparin at acidic pH indicates the existence of a distinct, lower affinity heparin-binding site in the protein. Chemical modification of the single histidine residue (His15) located in helix A of WT mGM-CSF with diethyl pyrocarbonate totally abolished binding to immobilized heparin. Moreover, replacement of His15 for an alanine residue significantly reduced the affinity of mGM-CSF for heparin at pH 5.0 and completely blocked heparin binding to a synthetic peptide corresponding to helix A of GM-CSF. These results indicate a major role of histidine residues in the regulation of the binding of GM-CSF to GAGs, supporting the notion that an acidic microenvironment is required for GM-CSF-dependent regulation of target cells. In addition, our results provide insight into the molecular basis of the strict species specificity of the biological activity of GM-CSF.
- Published
- 2005
- Full Text
- View/download PDF
22. Amyloidogenicity and Cytotoxicity of Recombinant Mature Human Islet Amyloid Polypeptide (rhIAPP)
- Author
-
Theri Leica Degaki, Mari Cleide Sogayar, Sergio T. Ferreira, Christian Colin, Adriano Sebollela, Marcelo N. N. Vieira, Ana Christina V. de Sousa, Ana Maria Blanco Martinez, Carlos Bloch, and Dahabada H. J. Lopes
- Subjects
Amyloid ,endocrine system ,DNA, Complementary ,Time Factors ,Recombinant Fusion Proteins ,Molecular Sequence Data ,Glycine ,Peptide ,Biochemistry ,law.invention ,Type ii diabetes ,law ,Escherichia coli ,Humans ,Amino Acid Sequence ,Cloning, Molecular ,Cytotoxicity ,Molecular Biology ,Cells, Cultured ,chemistry.chemical_classification ,geography ,geography.geographical_feature_category ,Base Sequence ,Dose-Response Relationship, Drug ,Reverse Transcriptase Polymerase Chain Reaction ,Chemistry ,Cell Biology ,Islet ,Recombinant Proteins ,Islet Amyloid Polypeptide ,Protein Structure, Tertiary ,Microscopy, Electron ,Spectrometry, Fluorescence ,Microscopy, Fluorescence ,Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization ,Mutation ,Recombinant DNA ,Electrophoresis, Polyacrylamide Gel ,Peptides - Abstract
Pancreatic amyloid plaques formed by the pancreatic islet amyloid polypeptide (IAPP) are present in more than 95% of type II diabetes mellitus patients, and their abundance correlates with the severity of the disease. IAPP is currently considered the most amyloidogenic peptide known, but the molecular bases of its aggregation are still incompletely understood. Detailed characterization of the mechanisms of amyloid formation requires large quantities of pure material. Thus, availability of recombinant IAPP in sufficient amounts for such studies constitutes an important step toward elucidation of the mechanisms of amyloidogenicity. Here, we report, for the first time, the successful expression, purification and characterization of the amyloidogenicity and cytotoxicity of recombinant human mature IAPP. This approach is likely to be useful for the production of other amyloidogenic peptides or proteins that are difficult to obtain by chemical synthesis.
- Published
- 2004
- Full Text
- View/download PDF
23. Acidic pH Modulates the Interaction between Human Granulocyte-Macrophage Colony-stimulating Factor and Glycosaminoglycans
- Author
-
Marcelo A. Carvalho, Silvia P. Azevedo, Sergio T. Ferreira, Aline Wettreich, Adriano Sebollela, Tatiana Coelho-Sampaio, and Radovan Borojevic
- Subjects
Models, Molecular ,Stromal cell ,Biochemistry ,Glycosaminoglycan ,Sulfation ,Glycolipid ,Naphthalenesulfonates ,Humans ,Molecular Biology ,Phospholipids ,Histidine ,Glycosaminoglycans ,chemistry.chemical_classification ,Binding Sites ,Heparin ,Sepharose ,Granulocyte-Macrophage Colony-Stimulating Factor ,Glycosidic bond ,Cell Biology ,Hydrogen-Ion Concentration ,Recombinant Proteins ,Spectrometry, Fluorescence ,Membrane ,chemistry ,Ionic strength ,Liposomes ,Protein Binding - Abstract
Granulocyte-macrophage colony-stimulating factor (GM-CSF) controls growth and differentiation of hematopoietic cells. Previous reports have indicated that the mitogenic activity of GM-CSF may be modulated by the glycosidic moiety of proteoglycans associated with the membrane of stromal cells. In this work, we have performed in vitro studies of the interaction between GM-CSF and glycosaminoglycans. The addition of heparin promoted a marked blue shift in the fluorescence emission spectrum of GM-CSF as well as a 30-fold increase in the intensity of light scattering, which indicates formation of large molecular weight complexes between the two molecules. Interestingly, heparin-induced changes in the spectral properties of GM-CSF were only observed at acidic pH. The dependence on acidic pH, together with a strict dependence on glycosaminoglycan sulfation and the fact that high ionic strength destabilized the interaction, indicates that the association between GM-CSF and glycosaminoglycans is mediated by electrostatic interactions. These interactions probably involve sulfate groups in the glycosaminoglycans and positively charged histidine residues in GM-CSF. We propose that negatively charged glycolipids present on the plasma membrane of the hematopoietic and/or the stromal cell could promote an acidic microenvironment capable of triggering interaction between GM-CSF and membrane-bound proteoglycans in vivo.
- Published
- 1999
- Full Text
- View/download PDF
24. Elucidating molecular mass and shape of a neurotoxic Aβ oligomer
- Author
-
Erika N. Cline, Kevin Luo, Kirsten L. Viola, William L. Klein, Adriano Sebollela, Pauline T. Velasco, Vinayak P. Dravid, Gina Mirela Mustata, Gajendra S. Shekhawat, and Kyle C. Wilcox
- Subjects
Physiology ,Microscopy, Atomic Force ,Hippocampus ,Biochemistry ,Oligomer ,Rats, Sprague-Dawley ,chemistry.chemical_compound ,0302 clinical medicine ,Pregnancy ,Immunotoxin ,neurotoxicity ,Cells, Cultured ,Neurons ,0303 health sciences ,Chemistry ,Immunotoxins ,SISTEMA NERVOSO CENTRAL ,Brain ,General Medicine ,Human brain ,3. Good health ,medicine.anatomical_structure ,Chromatography, Gel ,Female ,AFM ,Alzheimer's disease ,Alzheimer’s disease ,Research Article ,NU4 antibody ,medicine.drug_class ,Aβ oligomer ,Cognitive Neuroscience ,Monoclonal antibody ,Antibodies ,03 medical and health sciences ,Alzheimer Disease ,medicine ,Animals ,Humans ,030304 developmental biology ,Amyloid beta-Peptides ,Molecular mass ,Neurotoxicity ,Cell Biology ,Embryo, Mammalian ,medicine.disease ,In vitro ,Rats ,030217 neurology & neurosurgery - Abstract
Alzheimer's disease (AD), the most prevalent type of dementia, has been associated with the accumulation of amyloid β oligomers (AβOs) in the central nervous system. AβOs vary widely in size, ranging from dimers to larger than 100 kDa. Evidence indicates that not all oligomers are toxic, and there is yet no consensus on the size of the actual toxic oligomer. Here we used NU4, a conformation-dependent anti-AβO monoclonal antibody, to investigate size and shape of a toxic AβO assembly. By using size-exclusion chromatography and immuno-based detection, we isolated an AβO-NU4 complex amenable for biochemical and morphological studies. The apparent molecular mass of the NU4-targeted oligomer was 80 kDa. Atomic force microscopy imaging of the AβO-NU4 complex showed a size distribution centered at 5.37 nm, an increment of 1.5 nm compared to the size of AβOs (3.85 nm). This increment was compatible with the size of NU4 (1.3 nm), suggesting a 1:1 oligomer to NU4 ratio. NU4-reactive oligomers extracted from AD human brain concentrated in a molecular mass range similar to that found for in vitro prepared oligomers, supporting the relevance of the species herein studied. These results represent an important step toward understanding the connection between AβO size and toxicity.
- Published
- 2014
25. O3–02–03: Differential impact of high and low molecular mass beta‐amyloid oligomers on synapse density, receptor composition and memory in mice
- Author
-
Carine V Costa, Axa P Mota-Sales, William L. Klein, Cláudia P. Figueiredo, Jose Henrique Ledo, Julia R. Clarke, Sergio T. Ferreira, Helen M. Melo, Fernanda G. De Felice, Leonardo M. Saraiva, Felipe Ribeiro, and Adriano Sebollela
- Subjects
Molecular mass ,Amyloid ,Epidemiology ,Chemistry ,Health Policy ,Synapse ,Psychiatry and Mental health ,Cellular and Molecular Neuroscience ,Developmental Neuroscience ,Biophysics ,Composition (visual arts) ,Neurology (clinical) ,Geriatrics and Gerontology ,Receptor ,Beta (finance) ,Differential impact - Published
- 2013
- Full Text
- View/download PDF
26. Synapse-binding subpopulations of Aβ oligomers sensitive to peptide assembly blockers and scFv antibodies
- Author
-
Amanda L. Eckermann, Pauline T. Velasco, William L. Klein, Adriano Sebollela, Kevin B. Lee, Marie C. Heffern, Kirsten L. Viola, Izolda A. Popova, Benjamin N. Tiano, Pascale N. Lacor, Xiao-xia Sun, and Thomas J. Meade
- Subjects
Amyloid ,Physiology ,Immunoprecipitation ,Amyloid beta ,Cognitive Neuroscience ,Population ,Blotting, Western ,Peptide ,Biochemistry ,Oligomer ,chemistry.chemical_compound ,Humans ,education ,chemistry.chemical_classification ,education.field_of_study ,Amyloid beta-Peptides ,biology ,Cell Biology ,General Medicine ,Peptide Fragments ,Monomer ,chemistry ,Synapses ,biology.protein ,Electrophoresis, Polyacrylamide Gel ,Single-Chain Antibodies - Abstract
Amyloid β42 self-assembly is complex, with multiple pathways leading to large insoluble fibrils or soluble oligomers. Oligomers are now regarded as most germane to Alzheimer's pathogenesis. We have investigated the hypothesis that oligomer formation itself occurs through alternative pathways, with some leading to synapse-binding toxins. Immediately after adding synthetic peptide to buffer, solutions of Aβ42 were separated by a 50 kDa filter and fractions assessed by SDS-PAGE silver stain, Western blot, immunoprecipitation, and capacity for synaptic binding. Aβ42 rapidly assembled into aqueous-stable oligomers, with similar protein abundance in small (50 kDa) and large (50 kDa) oligomer fractions. Initially, both fractions were SDS-labile and resolved into tetramers, trimers, and monomers by SDS-PAGE. Upon continued incubation, the larger oligomers developed a small population of SDS-stable 10-16mers, and the smaller oligomers generated gel-impermeant complexes. The two fractions associated differently with neurons, with prominent synaptic binding limited to larger oligomers. Even within the family of larger oligomers, synaptic binding was associated with only a subset of these species, as a new scFv antibody (NUsc1) immunoprecipitated only a small portion of the oligomers while eliminating synaptic binding. Interestingly, low doses of the peptide KLVFFA blocked assembly of the 10-16mers, and this result was associated with loss of the smaller clusters of oligomers observed at synaptic sites. What distinguishes these smaller clusters from the unaffected larger clusters is not yet known. Results indicate that distinct species of Aβ oligomers are generated by alternative assembly pathways and that synapse-binding subpopulations of Aβ oligomers could be specifically targeted for Alzheimer's therapeutics.
- Published
- 2012
27. Amyloid-β oligomers induce differential gene expression in adult human brain slices
- Author
-
Samantha M. Martins, Sergio T. Ferreira, Helena Brentani, Fabio F. Oliveira, Léo Freitas-Correa, Dirce Maria Carraro, Fernanda G. De Felice, Soniza Vieira Alves-Leon, Andrea C. Paula-Lima, Leonardo M. Saraiva, Cesar Torres, Adriano Sebollela, Louise Danielle de Carvalho Mota, and Jorge Marcondes de Souza
- Subjects
Adult ,Male ,Transgene ,Cell ,Nerve Tissue Proteins ,Biology ,Biochemistry ,Neurobiology ,Alzheimer Disease ,Gene expression ,medicine ,Humans ,Cognitive decline ,Molecular Biology ,Oligonucleotide Array Sequence Analysis ,Regulation of gene expression ,Amyloid beta-Peptides ,Gene Expression Profiling ,Brain ,Cell Biology ,Actin cytoskeleton ,Cell biology ,Gene expression profiling ,medicine.anatomical_structure ,Gene Expression Regulation ,Synaptophysin ,biology.protein ,Female - Abstract
Cognitive decline in Alzheimer disease (AD) is increasingly attributed to the neuronal impact of soluble oligomers of the amyloid-β peptide (AβOs). Current knowledge on the molecular and cellular mechanisms underlying the toxicity of AβOs stems largely from rodent-derived cell/tissue culture experiments or from transgenic models of AD, which do not necessarily recapitulate the complexity of the human disease. Here, we used DNA microarray and RT-PCR to investigate changes in transcription in adult human cortical slices exposed to sublethal doses of AβOs. The results revealed a set of 27 genes that showed consistent differential expression upon exposure of slices from three different donors to AβOs. Functional classification of differentially expressed genes revealed that AβOs impact pathways important for neuronal physiology and known to be dysregulated in AD, including vesicle trafficking, cell adhesion, actin cytoskeleton dynamics, and insulin signaling. Most genes (70%) were down-regulated by AβO treatment, suggesting a predominantly inhibitory effect on the corresponding pathways. Significantly, AβOs induced down-regulation of synaptophysin, a presynaptic vesicle membrane protein, suggesting a mechanism by which oligomers cause synapse failure. The results provide insight into early mechanisms of pathogenesis of AD and suggest that the neuronal pathways affected by AβOs may be targets for the development of novel diagnostic or therapeutic approaches.
- Published
- 2012
28. Biological Targeting and Activity of Pre-fibrillar Aβ Assemblies
- Author
-
Jason Pitt, Adriano Sebollela, William L. Klein, Kyle C. Wilcox, Pascale N. Lacor, and Helen Martirosova
- Subjects
Synapse ,Programmed cell death ,Transduction (genetics) ,Insulin receptor ,biology ,Aβ oligomers ,biology.protein ,Aging brain ,Disease ,Neuroscience ,Intracellular - Abstract
The body of work from the past decade has advanced our understanding of how toxic oligomers of Aβ are capable of eliciting the spectrum of pathological and behavioral hallmarks of Alzheimer’s disease. These potent neurotoxins now provide a molecular basis for the cause of this disease as well as a basis for identifying and evaluating diagnostic and therapeutic strategies. Oligomer toxicity is mediated by a number of factors—both in the targeting of these toxins to the neuronal synapses and in the transduction of this targeting into intracellular signals resulting in synapse loss and, eventually, cell death. Recent investigations have focused on defining the mechanisms of binding of toxic Aβ oligomers, the pathways modulated by these events, and strategies to treat Alzheimer’s disease by targeting both aspects. One promising facet of recent research highlighted in this chapter, and in which Aβ oligomers play a central role, is the unfolding of connection between Alzheimer’s disease and insulin signaling in the aging brain.
- Published
- 2011
- Full Text
- View/download PDF
29. Soluble oligomers from a non-disease related protein mimic Abeta-induced tau hyperphosphorylation and neurodegeneration
- Author
-
Marcelo N N, Vieira, Letícia, Forny-Germano, Leonardo M, Saraiva, Adriano, Sebollela, Ana M Blanco, Martinez, Jean-Christophe, Houzel, Fernanda G, De Felice, and Sérgio T, Ferreira
- Subjects
Neurons ,Amyloid beta-Peptides ,Cell Survival ,Circular Dichroism ,Blotting, Western ,Fluorescent Antibody Technique ,tau Proteins ,Rats ,Rats, Sprague-Dawley ,Epitopes ,Microscopy, Electron ,Thiazoles ,Nephelometry and Turbidimetry ,Pregnancy ,Nerve Degeneration ,Chromatography, Gel ,Animals ,Electrophoresis, Polyacrylamide Gel ,Female ,Muramidase ,Benzothiazoles ,Phosphorylation ,Rats, Wistar ,Chickens ,Fluorescent Dyes - Abstract
Protein aggregation and amyloid accumulation in different tissues are associated with cellular dysfunction and toxicity in important human pathologies, including Alzheimer's disease and various forms of systemic amyloidosis. Soluble oligomers formed at the early stages of protein aggregation have been increasingly recognized as the main toxic species in amyloid diseases. To gain insight into the mechanisms of toxicity instigated by soluble protein oligomers, we have investigated the aggregation of hen egg white lysozyme (HEWL), a normally harmless protein. HEWL initially aggregates into beta-sheet rich, roughly spherical oligomers which appear to convert with time into protofibrils and mature amyloid fibrils. HEWL oligomers are potently neurotoxic to rat cortical neurons in culture, while mature amyloid fibrils are little or non-toxic. Interestingly, when added to cortical neuronal cultures HEWL oligomers induce tau hyperphosphorylation at epitopes that are characteristically phosphorylated in neurons exposed to soluble oligomers of the amyloid-beta peptide. Furthermore, injection of HEWL oligomers in the cerebral cortices of adult rats induces extensive neurodegeneration in different brain areas. These results show that soluble oligomers from a non-disease related protein can mimic specific neuronal pathologies thought to be induced by soluble amyloid-beta peptide oligomers in Alzheimer's disease and support the notion that amyloid oligomers from different proteins may share common structural determinants that would explain their generic cytotoxicities.
- Published
- 2007
30. Heparin-binding sites in granulocyte-macrophage colony-stimulating factor. Localization and regulation by histidine ionization
- Author
-
Adriano, Sebollela, Thiago C, Cagliari, Gabriel S C S, Limaverde, Alex, Chapeaurouge, Marcos H F, Sorgine, Tatiana, Coelho-Sampaio, Carlos H I, Ramos, and Sérgio T, Ferreira
- Subjects
Binding Sites ,Heparin ,Phosphorylcholine ,Molecular Sequence Data ,Granulocyte-Macrophage Colony-Stimulating Factor ,Protein Structure, Secondary ,Protein Structure, Tertiary ,Mice ,Species Specificity ,Mutation ,Animals ,Humans ,Histidine ,Amino Acid Sequence ,Heparitin Sulfate ,Peptides - Abstract
The biological activity of granulocyte-macrophage colony-stimulating factor (GM-CSF) is modulated by the sulfated glycosaminoglycans (GAGs) heparan sulfate and heparin. However, the molecular mechanisms involved in such interactions are still not completely understood. We have proposed previously that helix C, one of the four alpha-helices of human GM-CSF (hGM-CSF), contains a GAG-binding site in which positively charged residues are spatially positioned for interaction with the sulfate moieties of the GAGs (Wettreich, A., Sebollela, A., Carvalho, M. A., Azevedo, S. P., Borojevic, R., Ferreira, S. T., and Coelho-Sampaio, T. (1999) J. Biol. Chem. 274, 31468-31475). Protonation of two histidine residues (His83 and His87) in helix C of hGM-CSF appears to act as a pH-dependent molecular switch to control the interaction with GAGs. Based on these findings, we have now generated a triple mutant form of murine GM-CSF (mGM-CSF) in which three noncharged residues in helix C of the murine factor (Tyr83, Gln85, and Tyr87) were replaced by the corresponding basic residues present in hGM-CSF (His83, Lys85, and His87). Binding assays on heparin-Sepharose showed that, at acidic pH, the triple mutant mGM-CSF binds to immobilized heparin with significantly higher affinity than wild type (WT) mGM-CSF and that neither protein binds to the column at neutral pH. The fact that even WT mGM-CSF binds to heparin at acidic pH indicates the existence of a distinct, lower affinity heparin-binding site in the protein. Chemical modification of the single histidine residue (His15) located in helix A of WT mGM-CSF with diethyl pyrocarbonate totally abolished binding to immobilized heparin. Moreover, replacement of His15 for an alanine residue significantly reduced the affinity of mGM-CSF for heparin at pH 5.0 and completely blocked heparin binding to a synthetic peptide corresponding to helix A of GM-CSF. These results indicate a major role of histidine residues in the regulation of the binding of GM-CSF to GAGs, supporting the notion that an acidic microenvironment is required for GM-CSF-dependent regulation of target cells. In addition, our results provide insight into the molecular basis of the strict species specificity of the biological activity of GM-CSF.
- Published
- 2005
31. Inhibition of yeast glutathione reductase by trehalose: possible implications in yeast survival and recovery from stress
- Author
-
Adriano Sebollela, Tatiana Coelho-Sampaio, Paulo Roberto Louzada, Verietta Sarone-Williams, Sergio T. Ferreira, and Mauro Sola-Penna
- Subjects
Protein Folding ,Sucrose ,Antioxidant ,Hot Temperature ,medicine.medical_treatment ,Saccharomyces cerevisiae ,Glutathione reductase ,Glucosephosphate Dehydrogenase ,Biochemistry ,chemistry.chemical_compound ,Osmotic Pressure ,medicine ,Trehalase ,Pyrophosphatases ,chemistry.chemical_classification ,biology ,Ethanol ,Spectrum Analysis ,Trehalose ,Cell Biology ,biology.organism_classification ,Yeast ,Betaine ,Cytosol ,Enzyme ,Glutathione Reductase ,chemistry - Abstract
Accumulation of trehalose has been implicated in the tolerance of yeast cells to several forms of stress, including heat-shock and high ethanol levels. However, yeast lacking trehalase, the enzyme that degrades trehalose, exhibit poor survival after exposure to stress conditions. This suggests that optimal cell viability also depends on the capacity to rapidly degrade the high levels of trehalose that build up under stress. Here, we initially examined the effects of trehalose on the activity of an important antioxidant enzyme, glutathione reductase (GR), from Saccharomyces cerevisiae . At 25 °C, GR was inhibited by trehalose in a dose-dependent manner, with 70% inhibition at 1.5 M trehalose. The inhibition was practically abolished at 40 °C, a temperature that induces a physiological response of trehalose accumulation in yeast. The inhibition of GR by trehalose was additive to the inhibition caused by ethanol, indicating that enzyme function is drastically affected upon ethanol-induced stress. Moreover, two other yeast enzymes, cytosolic pyrophosphatase and glucose 6-phosphate dehydrogenase, showed temperature dependences on inhibition by trehalose that were similar to the temperature dependence of GR inhibition. These results are discussed in terms of the apparent paradox represented by the induction of enzymes involved in both synthesis and degradation of trehalose under stress, and suggest that the persistence of high levels of trehalose after recovery from stress could lead to the inactivation of important yeast enzymes.
- Published
- 2003
32. Amyloid β-Peptide Oligomers Stimulate RyR-Mediated Ca2+Release Inducing Mitochondrial Fragmentation in Hippocampal Neurons and Prevent RyR-Mediated Dendritic Spine Remodeling Produced by BDNF.
- Author
-
Andrea C. Paula-Lima, Tatiana Adasme, Carol SanMartín, Adriano Sebollela, Claudio Hetz, M. Angélica Carrasco, Sergio T. Ferreira, and Cecilia Hidalgo
- Published
- 2011
- Full Text
- View/download PDF
33. Predissociated Dimers and Molten Globule Monomers in the Equilibrium Unfolding of Yeast Glutathione Reductase
- Author
-
Paulo Roberto Louzada, Adriano Sebollela, Sergio T. Ferreira, and Marcelo E. Scaramello
- Subjects
Circular dichroism ,Protein Folding ,Hot Temperature ,Protein Conformation ,Dimer ,Equilibrium unfolding ,Biophysics ,Saccharomyces cerevisiae ,Protein Structure, Secondary ,chemistry.chemical_compound ,Enzyme Stability ,Organic chemistry ,Guanidine ,Quenching (fluorescence) ,Circular Dichroism ,Proteins ,Molten globule ,Enzyme Activation ,Crystallography ,Glutathione Reductase ,Spectrometry, Fluorescence ,chemistry ,Protein quaternary structure ,Protein folding ,Dimerization - Abstract
The equilibrium unfolding of dimeric yeast glutathione reductase (GR) by guanidine hydrochloride (GdnHCl) was investigated. Unfolding was monitored by a variety of techniques, including intrinsic fluorescence emission, anisotropy and iodide quenching measurements, far-ultraviolet circular dichroism and thiol reactivity measurements. At 1M GdnHCl, one thiol group of GR became accessible to modification with 5,5′-dithiobis-(2-nitrobenzoic) acid (DTNB), whereas no changes could be detected in the spectroscopic properties (fluorescence, circular dichroism) of the protein. Between 2 and 3M GdnHCl, two partially folded intermediate states possessing flexible tertiary structures (revealed by fluorescence data) but compact secondary structures (as indicated by circular dichroism measurements) were identified. The quaternary structure of GR in the presence of GdnHCl was also investigated by size-exclusion liquid chromatography. These results indicated the presence of an expanded predissociated dimer at 2.5MGdnHCl and partially folded monomers at 3M GdnHCl. Taken together, these results suggest the existence of two molten-globule-like intermediate species (one dimeric and one monomeric) in the unfolding of GR. The results are discussed in terms of the mechanism of GR folding and dimerization.
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.