Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Maria Grazia Spillantini is active.

Publication


Featured researches published by Maria Grazia Spillantini.


Neuron | 1989

Multiple isoforms of human microtubule-associated protein tau: sequences and localization in neurofibrillary tangles of Alzheimer's disease

Michel Goedert; Maria Grazia Spillantini; Ross Jakes; D. Rutherford; R.A. Crowther

We have determined the sequences of isoforms of human tau protein, which differ from previously reported forms by insertions of 29 or 58 amino acids in the amino-terminal region. Complementary DNA cloning shows that the insertions occur in combination with both three and four tandem repeats. RNAase protection assays indicate that transcripts encoding isoforms with the insertions are expressed in an adult-specific manner. Transcripts encoding four tandem repeats are also expressed in an adult-specific manner, whereas mRNAs encoding three tandem repeats are expressed throughout life, including in fetal brain. The levels of transcripts encoding the 29 or 58 amino acid inserts were not significantly changed in cerebral cortex from patients with Alzheimers disease. Antisera raised against synthetic peptides corresponding to these different human tau isoforms demonstrate that multiple tau protein isoforms are incorporated into the neurofibrillary tangles of Alzheimers disease.


Neuron | 1992

Tau proteins of alzheimer paired helical filaments: Abnormal phosphorylation of all six brain isoforms

Michel Goedert; Maria Grazia Spillantini; Nigel J. Cairns; R.A. Crowther

Preparations of dispersed paired helical filaments (PHFs) from the brains of Alzheimers disease and Downs syndrome patients display on gels three principal bands corresponding to abnormally modified forms of the microtubule-associated protein tau. Interpretation of the pattern is difficult because there are six tau isoforms in normal brain and phosphorylation changes their mobility. By enzymatic dephosphorylation at high temperature, we have shifted the three abnormal bands obtained from dispersed PHFs to align with the six nonphosphorylated tau isoforms. By using antibodies specific for some of the inserts that distinguish the various isoforms and label PHFs, we have established a correspondence between PHFs, abnormal bands, and isoforms. This identification of isoforms is a necessary step in unravelling the molecular pathogenesis of PHFs.


FEBS Letters | 1994

Identification of two distinct synucleins from human brain

Ross Jakes; Maria Grazia Spillantini; Michel Goedert

Two abundant proteins of 140 and 134 amino acids were purified and sequenced from human brain. They were identified through their reactivity on immunoblots with a partially characterised monoclonal antibody that recognises tau protein in a phosphorylation‐dependent manner. The 140 amino acid protein is identical with the precursor of the non‐Aβ component of Alzheimers disease amyloid which in turn is highly homologous to synuclein from Torpedo electroplaques and rat brain. The 134 amino acid protein is the human homologue of bovine phosphoneuroprotein 14; it is 61% identical in sequence to the 140 amino acid protein. The previously unrecognised homology between these two proteins defines a family of human brain synucleins. We refer to the 140 and 134 amino acid proteins as α‐synuclein and β‐synuclein, respectively. Both synucleins are expressed predominantly in brain, where they are concentrated in presynaptic nerve terminals.


The EMBO Journal | 1989

Cloning and sequencing of the cDNA encoding an isoform of microtubule-associated protein tau containing four tandem repeats: differential expression of tau protein mRNAs in human brain.

Michel Goedert; Maria Grazia Spillantini; M.-C. Potier; J. Ulrich; R.A. Crowther

We have isolated cDNA clones encoding a 383‐amino acid isoform of the human microtubule‐associated protein tau. It differs from previously determined tau sequences by the presence of an additional repeat of 31 amino acids, giving four, rather than three, tandem repeats in its carboxy‐terminal half. The extra repeat is encoded by a separate exon. Probes derived from cDNA clones encoding the three (type I) and four repeat (type II) tau protein isoforms detected mRNAs for both forms in all adult human brain areas examined. However, in foetal brain only type I mRNA was found. Type I and type II mRNAs were present in pyramidal cells in cerebral cortex. In the hippocampal formation, type I mRNA was found in pyramidal and granule cells; type II mRNA was detected in most, though not all, pyramidal cells but not in granule cells. These observations indicate that tau protein mRNAs are expressed in a stage‐ and cell‐specific manner. Tau protein is found in the protease‐resistant core of the paired helical filament, the major constituent of the neurofibrillary tangle in Alzheimers disease. Taken in conjunction with previous findings, the present results indicate that both the three and four repeat‐containing tau protein isoforms are present in the core of the paired helical filament.


The EMBO Journal | 1995

Somatodendritic localization and hyperphosphorylation of tau protein in transgenic mice expressing the longest human brain tau isoform.

Jürgen Götz; Alphonse Probst; Maria Grazia Spillantini; T Schäfer; Ross Jakes; Kurt Bürki; Michel Goedert

Microtubule‐associated protein tau is the major constituent of the paired helical filament, the main fibrous component of the neurofibrillary lesions of Alzheimers disease. Tau is an axonal phosphoprotein in normal adult brain. In Alzheimers disease brain tau is hyperphosphorylated and is found not only in axons, but also in cell bodies and dendrites of affected nerve cells. We report the production and analysis of transgenic mice that express the longest human brain tau isoform under the control of the human Thy‐1 promoter. As in Alzheimers disease, transgenic human tau protein was present in nerve cell bodies, axons and dendrites; moreover, it was phosphorylated at sites that are hyperphosphorylated in paired helical filaments. We conclude that transgenic human tau protein showed pre‐tangle changes similar to those that precede the full neurofibrillary pathology in Alzheimers disease.


Acta Neuropathologica | 1996

Pick’s disease: hyperphosphorylated tau protein segregates to the somatoaxonal compartment

Alphonse Probst; Markus Tolnay; D. Langui; Michel Goedert; Maria Grazia Spillantini

Abstract Pick bodies and ballooned cells of Pick’s disease and the neurofibrillary lesions of Alzheimer’s disease are characterized by the presence of hyperphosphorylated microtubule-associated protein tau. Little is known about the mechanisms underlying tau hyperphosphorylation in Pick’s disease and the distribution of abnormal tau in affected neurons. We have used a panel of phosphorylation-dependent (AT270, AT8, AT180, 12E8, PHF-1, AT10 and Tau-1) and phosphorylation-independent anti-tau antibodies (N-tau 5 and 134) to stain brain tissue sections from subjects with Pick’s disease and Alzheimer’s disease. These antibodies labeled Pick bodies and neurofibrillary lesions in a similar way, with the exception of antibody 12E8, which stained a subset of neurofibrillary tangles, but no Pick bodies. Moreover, abundant AT8- and PHF-1-positive neuritic profiles were observed in cortical areas rich in Pick bodies, even in the complete absence of neurofibrillary lesions. Unlike the Gallyas-positive neuropil threads of Alzheimer’s disease, which were of variable diameter and covered by spiny appendages, neuritic profiles of Pick’s disease showed a regular diameter, appeared smooth and were Gallyas-negative. In contrast to Alzheimer’s disease, dendritic branches of neurons containing Pick bodies were not labeled by anti-tau antibodies. In the hippocampus, numerous tau-positive axon terminals were found along dendrites of the polymorphic layer of the dentate gyrus. Our results indicate that tau proteins in Pick’s disease and Alzheimer’s disease share similar phosphorylated residues, with the exception of serine 262, which is phosphorylated in Alzheimer tangles but not in Pick bodies or neuritic profiles. Furthermore, we show that hyperphosphorylated tau segregates to different neuronal compartments in the two diseases, with a somatoaxonal distribution in Pick’s disease and a somatodendritic distribution in Alzheimer’s disease.


Journal of Neuropathology and Experimental Neurology | 1999

Tau pathology in a family with dementia and a P301L mutation in tau

Suzanne S. Mirra; Jill R. Murrell; Marla Gearing; Maria Grazia Spillantini; Michel Goedert; R. Anthony Crowther; Allan I. Levey; Randi Jones; Joanne Green; John M. Shoffner; Bruce H. Wainer; M. L. Schmidt; John Q. Trojanowski; Bernardino Ghetti

Familial forms of frontotemporal dementia and parkinsonism linked to chromosome 17 (FTDP-17) have recently been associated with coding region and intronic mutations in the tau gene. Here we report our findings on 2 affected siblings from a family with early-onset dementia, characterized by extensive tau pathology and a Pro to Leu mutation at codon 301 of tau. The proband, a 55-year-old woman, and her 63-year-old brother died after a progressive dementing illness clinically diagnosed as Alzheimer disease. Their mother, 2 sisters, maternal aunt and uncle, and several cousins were also affected. Autopsy in both cases revealed frontotemporal atrophy and degeneration of basal ganglia and substantia nigra. Sequencing of exon 10 of the tau gene revealed a C to T transition at codon 301, resulting in a Pro to Leu substitution. Widespread neuronal and glial inclusions, neuropil threads, and astrocytic plaques similar to those seen in corticobasal degeneration were labeled with a battery of antibodies to phosphorylation-dependent and phosphorylation-independent epitopes spanning the entire tau sequence. Isolated tau filaments had the morphology of narrow twisted ribbons. Sarkosyl-insoluble tau exhibited 2 major bands of 64 and 68 kDa and a minor 72 kDa band, similar to the pattern seen in a familial tauopathy associated with an intronic tau mutation. These pathological tau bands predominantly contained the subset of tau isoforms with 4 microtubule-binding repeats selectively affected by the P301L missense mutation. Our findings emphasize the phenotypic and genetic heterogeneity of tauopathies and highlight intriguing links between FTDP-17 and other neurodegenerative diseases.


Journal of Biological Chemistry | 1999

Stress-activated Protein Kinase-3 Interacts with the PDZ Domain of α1-Syntrophin A MECHANISM FOR SPECIFIC SUBSTRATE RECOGNITION

Masato Hasegawa; Ana Cuenda; Maria Grazia Spillantini; Gareth M. Thomas; Valérie Buée-Scherrer; Philip Cohen; Michel Goedert

Mechanisms for selective targeting to unique subcellular sites play an important role in determining the substrate specificities of protein kinases. Here we show that stress-activated protein kinase-3 (SAPK3, also called ERK6 and p38γ), a member of the mitogen-activated protein kinase family that is abundantly expressed in skeletal muscle, binds through its carboxyl-terminal sequence -KETXL to the PDZ domain of α1-syntrophin. SAPK3 phosphorylates α1-syntrophin at serine residues 193 and 201 in vitro and phosphorylation is dependent on binding to the PDZ domain of α1-syntrophin. In skeletal muscle SAPK3 and α1-syntrophin co-localize at the neuromuscular junction, and both proteins can be co-immunoprecipitated from transfected COS cell lysates. Phosphorylation of a PDZ domain-containing protein by an associated protein kinase is a novel mechanism for determining both the localization and the substrate specificity of a protein kinase.


Acta Neuropathologica | 1996

Comparison of the neurofibrillary pathology in Alzheimer’s disease and familial presenile dementia with tangles

Maria Grazia Spillantini; R.A. Crowther; Michel Goedert

Abstract Alzheimer’s disease (AD) is characterised neuropathologically by the presence of abundant extracellular β-amyloid deposits and intracellular neurofibrillary lesions consisting of neurofibrillary tangles, neuropil threads and senile plaque neurites which contain paired helical filaments (PHFs) made of hyperphosphorylated microtubule-associated protein tau. A new familial form of presenile dementia with neurofibrillary pathology and no β-amyloid deposits has been described recently [Sumi et al. (1992) Neurology 42: 120–127]. We have compared the tau pathology in this familial form of presenile dementia with that of AD. To this end we have used electron microscopy, immunoblotting and immunohistochemistry with phosphorylation-dependent (PHF1, AT8, AT100, AT180, AT270, 12E8) and phosphorylation-independent (BR133, BR134) anti-tau antibodies. We show that in the two diseases dispersed PHFs are structurally, biochemically and immunologically identical; they are stained by all anti-tau antibodies used and on immunoblots PHF-tau appears as three major bands of 60, 64 and 68 kDa. However, while the anti-tau antibodies stain neurofibrillary tangles, neuropil threads and neuritic plaques in AD brain, no neuritic plaques are found in familial presenile dementia. These results indicate that in the two diseases tau undergoes the same modifications; they confirm that neurofibrillary tangles and neuropil threads like those in AD can exist independently of β-amyloid deposits and that their presence is associated with dementia.


Brain Pathology | 1991

Tau Proteins and Neurofibrillary Degeneration

Michel Goedert; Maria Grazia Spillantini; R.A. Crowther

The paired helical filament is the major fibrous component of neurofibrillary pathology in Alzheimers disease. Over the last three years evidence has accumulated that the microtubule‐associated protein tau forms an important, if not the sole, constituent of the paired helical filament. Tau protein in normal brain is bound to axonal microtubules by a tandem repeat region. In Alzheimers disease a proportion of tau protein becomes abnormally phos‐phorylated and is no longer associated with axonal microtubules but instead accumulates in paired helical filaments throughout affected nerve cells. The tandem repeat region contributes substantially to the structural core of the paired helical filament, around which the amino‐terminal half of the molecule forms a disordered coat.

Collaboration


Dive into the Maria Grazia Spillantini's collaboration.

Top Co-Authors

Avatar

Michel Goedert

Laboratory of Molecular Biology

View shared research outputs
Top Co-Authors

Avatar

Ross Jakes

Laboratory of Molecular Biology

View shared research outputs
Top Co-Authors

Avatar

R.A. Crowther

Laboratory of Molecular Biology

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Aaron Klug

Medical Research Council

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Michael J. Smith

Laboratory of Molecular Biology

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge