Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
1.
Prion ; 16(1): 66-77, 2022 12.
Article in English | MEDLINE | ID: mdl-35737759

ABSTRACT

Engaging patients as partners in biomedical research has gradually gained consensus over the last two decades. They provide a different perspective on health priorities and help to improve design and outcomes of clinical studies. This paper describes the relationship established between scientists and members of a large family at genetic risk of very rare lethal disease, fatal familial insomnia (FFI). This interaction led to a clinical trial based on the repurposing of doxycycline - an antibiotic with a known safety profile and optimal blood-brain barrier passage - which in numerous preclinical and clinical studies had given evidence of its potential therapeutic effect in neurodegenerative disorders, including prion diseases like FFI. The design of this trial posed several challenges, which were addressed jointly by the scientists and the FFI family. Potential participants excluded the possibility of being informed of their own FFI genotype; thus, the trial design had to include both carriers of the FFI mutation (10 subjects), and non-carriers (15 subjects), who were given placebo. Periodic clinical controls were performed on both groups by blinded examiners. The lack of surrogate outcome measures of treatment efficacy has required to compare the incidence of the disease in the treated group with a historical dataset during 10 years of observation. The trial is expected to end in 2023. Regardless of the clinical outcome, it will provide worthwhile knowledge on the disease. It also offers an important example of public engagement and collaboration to improve the quality of clinical science.


Subject(s)
Insomnia, Fatal Familial , Prion Diseases , Humans , Insomnia, Fatal Familial/drug therapy , Insomnia, Fatal Familial/genetics , Mutation , Prion Diseases/genetics
2.
J Biol Chem ; 296: 100490, 2021.
Article in English | MEDLINE | ID: mdl-33662396

ABSTRACT

Fatal familial insomnia (FFI), genetic Creutzfeldt-Jakob disease (gCJD), and Gerstmann-Sträussler-Scheinker (GSS) syndrome are neurodegenerative disorders linked to prion protein (PrP) mutations. The pathogenic mechanisms are not known, but increasing evidence points to mutant PrP misfolding and retention in the secretory pathway. We previously found that the D178N/M129 mutation associated with FFI accumulates in the Golgi of neuronal cells, impairing post-Golgi trafficking. In this study we further characterized the trafficking defect induced by the FFI mutation and tested the 178N/V129 variant linked to gCJD and a nine-octapeptide repeat insertion associated with GSS. We used transfected HeLa cells, embryonic fibroblasts and primary neurons from transgenic mice, and fibroblasts from carriers of the FFI mutation. In all these cell types, the mutant PrPs showed abnormal intracellular localizations, accumulating in the endoplasmic reticulum (ER) and Golgi. To test the efficiency of the membrane trafficking system, we monitored the intracellular transport of the temperature-sensitive vesicular stomatite virus glycoprotein (VSV-G), a well-established cargo reporter, and of endogenous procollagen I (PC-I). We observed marked alterations in secretory trafficking, with VSV-G accumulating mainly in the Golgi complex and PC-I in the ER and Golgi. A redacted version of mutant PrP with reduced propensity to misfold did not impair VSV-G trafficking, nor did artificial ER or Golgi retention of wild-type PrP; this indicates that both misfolding and intracellular retention were required to induce the transport defect. Pharmacological activation of Src family kinase (SFK) improved intracellular transport, suggesting that mutant PrP impairs secretory trafficking through corruption of SFK-mediated signaling.


Subject(s)
Mutation , Prion Proteins/metabolism , src-Family Kinases/metabolism , Animals , Cells, Cultured , Creutzfeldt-Jakob Syndrome/genetics , Creutzfeldt-Jakob Syndrome/metabolism , Creutzfeldt-Jakob Syndrome/pathology , Disease Models, Animal , Endoplasmic Reticulum/metabolism , Enzyme Activation , Gerstmann-Straussler-Scheinker Disease/genetics , Gerstmann-Straussler-Scheinker Disease/metabolism , Gerstmann-Straussler-Scheinker Disease/pathology , Golgi Apparatus/metabolism , Humans , Insomnia, Fatal Familial/genetics , Insomnia, Fatal Familial/metabolism , Insomnia, Fatal Familial/pathology , Mice , Mice, Inbred C57BL , Mice, Inbred CBA , Mice, Transgenic , Prion Proteins/genetics , Protein Folding , Secretory Pathway , src-Family Kinases/chemistry
3.
Mol Neurobiol ; 57(4): 1889-1903, 2020 Apr.
Article in English | MEDLINE | ID: mdl-31879858

ABSTRACT

Mechanisms of tissue damage in Huntington's disease involve excitotoxicity, mitochondrial damage, and inflammation, including microglia activation. Immunomodulatory and anti-protein aggregation properties of tetracyclines were demonstrated in several disease models. In the present study, the neuroprotective and anti-inflammatory effects of the tetracycline doxycycline were investigated in the mouse model of HD disease R6/2. Transgenic mice were daily treated with doxycycline 20 mg/kg, starting from 4 weeks of age. After sacrifice, histological and immunohistochemical studies were performed. We found that doxycycline-treated R6/2 mice survived longer and displayed less severe signs of neurological dysfunction than the saline-treated ones. Primary outcome measures such as striatal atrophy, neuronal intranuclear inclusions, and the negative modulation of microglial reaction revealed a neuroprotective effect of the compound. Doxycycline provided a significantly increase of activated CREB and BDNF in the striatal neurons, along with a down modulation of neuroinflammation, which, combined, might explain the beneficial effects observed in this model. Our findings show that doxycycline treatment could be considered as a valid therapeutic approach for HD.


Subject(s)
Doxycycline/therapeutic use , Huntington Disease/drug therapy , Neuroprotective Agents/therapeutic use , Animals , Behavior, Animal/drug effects , Brain-Derived Neurotrophic Factor/metabolism , Corpus Striatum/drug effects , Corpus Striatum/pathology , Corpus Striatum/physiopathology , Cyclic AMP Response Element-Binding Protein/metabolism , Disease Models, Animal , Disks Large Homolog 4 Protein/metabolism , Doxycycline/pharmacology , Female , Huntington Disease/physiopathology , Male , Mice, Transgenic , Microglia/drug effects , Microglia/metabolism , Motor Activity/drug effects , Neurons/drug effects , Neurons/pathology , Neuroprotective Agents/pharmacology , Open Field Test , Organ Size/drug effects , Survival Analysis , Weight Loss/drug effects
4.
Curr Alzheimer Res ; 16(1): 1-7, 2019.
Article in English | MEDLINE | ID: mdl-30381075

ABSTRACT

BACKGROUND: Presenilin-1 (PSEN-1) is a component of the γ-secretase complex involved in ß-amyloid Precursor Protein (AßPP) processing. Usually, Alzheimer's disease (AD)-linked mutations in the PSEN-1 gene lead to the early onset and increase the production of the aggregation-prone peptide Aß42. However, the PSEN-1 E318G variant has an unclear pathogenic role and is recently reported as a genetic risk factor for AD. In particular, E318G variant presence correlated with increased cerebrospinal fluid (CSF) levels of Total Tau (t-tau) and Phosphorylated Tau (p-tau). OBJECTIVE: We describe a large Italian family, which we followed from January 2003 to January 2018, with the late-onset AD and the E318G variant, with the aim of assessing E318G-associated CSF or plasma biochemical changes in biomarkers of dementia. METHOD: CSF Aß42, t-tau and p-tau, plasma Aß42 and Aß40 were assessed by ELISA tests, while CSF amyloid peptides profile was investigated by mass spectrometry. RESULTS: We did not find any changes in CSF biochemical markers (Aß42, t-tau, p-tau and amyloid peptides) of asymptomatic E318G carriers in 2010 and 2012, but plasma Aß40 was increased at the same times. From 2003 to 2018, no asymptomatic E318G carrier developed AD. CONCLUSION: Our follow-up of this family may help elucidate E318G's role in AD and globally points to a null effect of this variant.


Subject(s)
Dementia/cerebrospinal fluid , Dementia/genetics , Presenilin-1/genetics , Age of Onset , Biomarkers/blood , Biomarkers/cerebrospinal fluid , Dementia/blood , Dementia/epidemiology , Family , Female , Follow-Up Studies , Genetic Predisposition to Disease , Genetic Variation , Heterozygote , Humans , Male , Middle Aged
5.
Mov Disord ; 31(6): 771-81, 2016 06.
Article in English | MEDLINE | ID: mdl-27030592

ABSTRACT

The term oligomeropathies defines the neurodegenerative disorders associated with protein misfolding, where small soluble aggregates (oligomers 4-200 KDa) are the cause of neuronal dysfunction and are responsible for spreading the pathology. The ability of these soluble ß-sheet conformers to induce neuronal damage has been investigated in direct challenge with the monomeric and fibrillary structures, showing that only the oligomeric species affected the neurons. ß amyloid oligomers were initially purified from Alzheimer brains and obtained using synthetic peptides. Together with the neuronal death, synaptic dysfunction, loss of spines, and LTP impairment were seen with the direct application of ß amyloid oligomers. Similar results have been described with proteins associated with other neurodegenerative disorders. The biological activities of oligomeric forms of α synuclein have been described in Parkinson's disease and Lewy body dementia. Detrimental effects have been associated with the oligomeric forms of prion, tau, and huntingtin, the key proteins in prion diseases, frontotemporal dementia, and Huntington's disease, respectively. The molecular mechanisms of the oligomer-related toxic effects can be summarized under three headings: nonspecific perturbance of cellular and intracellular membranes, specific interaction with various cellular entities, and amyloid pore channel formation. To characterize and distinguish oligomer actions better, we compared the ability of ß amyloid and α synuclein oligomers to induce cognitive impairment when applied directly into the brain in the same acute mouse model. We also investigated the role of inflammatory components. © 2016 International Parkinson and Movement Disorder Society.


Subject(s)
Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Parkinson Disease/metabolism , Proteostasis Deficiencies/metabolism , alpha-Synuclein/metabolism , Animals , Humans
6.
Prion ; 9(2): 75-9, 2015.
Article in English | MEDLINE | ID: mdl-25996399

ABSTRACT

The text describes a preventive clinical trial with drug treatment in a very rare neurodegenerative disease (Fatal familial Insomnia, FFI) designed with the help of individuals at genetic risk of developing the disease, asymptomatic carriers, who have agreed to be exposed over a 10-year period to doxycycline, an antibiotic with anti-prion activity. At least 10 carriers of the FFI mutation over 42 y old will be treated with doxycycline (100 mg/die) and the incidence of the disease will be compared to that of an historical dataset. For ethical reasons a randomized, double-blind, placebo-controlled trial was not feasible, however the study design and the statistical analysis ensure the scientific value of the results. This approach might represent an important breakthrough in terms of potential therapy and knowledge of rare diseases that could give some hopes to these neglected patients.


Subject(s)
Insomnia, Fatal Familial , Adult , Clinical Trials as Topic , Doxycycline/therapeutic use , Humans , Insomnia, Fatal Familial/drug therapy , Insomnia, Fatal Familial/genetics , Insomnia, Fatal Familial/prevention & control , Middle Aged
7.
Curr Top Med Chem ; 13(19): 2465-76, 2013.
Article in English | MEDLINE | ID: mdl-24059336

ABSTRACT

In the last two decades, knowledge of the neurobiology of prion diseases or transmissible spongiform encephalopathies (TSE) has significantly advanced, but a successful therapy to stop or delay the progression of these disorders remains one of the most challenging goals of biomedical research. Several obstacles to this achievement are in common with other neurodegenerative disorders: difficulties to move from experimental level to clinical stage; appropriate timing of intervention; correct set up of clinical trial. Also in terms of molecular bases of disease, TSE and the other neurodegenerative disorders associated with protein misfolding such as Alzheimer, Parkinson and Huntington diseases, share a central pathogenic role of soluble small aggregates, named oligomers, considered the culprit of neuronal dysfunction: accordingly, these disorders could by termed oligomeropathies. However, the rapid progression of TSE, together with their clinical and molecular heterogeneity, make the therapeutic approach particularly problematic. The main target of the antiprion strategy has been the pathological form of the cellular prion protein (PrP(C)) termed PrP(Sc), invariably associated with the diseases. Several compounds have been found to affect PrP(Sc) formation or enhance its clearance in in vitro models, and prolong survival in experimental animals. However, few of them such as quinacrine and pentosan polysulfate have reached the clinical evaluation; more recently, we have conducted a clinical trial with doxycycline in patients with Creutzfeldt-Jakob disease without satisfactory results. In experimental conditions, active and passive immunization with antibodies against PrP and mucosal vaccination have shown to protect from peripheral infection. Other studies have proposed new potentially effective molecules targeting PrP oligomers. Furthermore, the possibility to interfere with PrP(C) to PrP(Sc) conversion by an active control of PrP(C) is another interesting approach emerging from experimental studies. However, in common with the other oligomeropathies, early diagnosis allowing to treat at risk population in a preclinical stage represent the more realistic perspective for efficient TSE therapy.


Subject(s)
Doxycycline/therapeutic use , Pentosan Sulfuric Polyester/therapeutic use , Prion Diseases/drug therapy , Quinacrine/therapeutic use , Animals , Doxycycline/pharmacology , Humans , Pentosan Sulfuric Polyester/pharmacology , Prion Diseases/diagnosis , Prions/antagonists & inhibitors , Prions/pathogenicity , Quinacrine/pharmacology
8.
Neurobiol Aging ; 28(11): 1682-8, 2007 Nov.
Article in English | MEDLINE | ID: mdl-16952411

ABSTRACT

Presenilin-1 (PSEN-1) is a component of the gamma-secretase complex involved in beta-amyloid precursor protein (betaAPP) processing. To date about 140 pathogenic mutations in the PSEN-1 gene have been identified and their main biochemical effect is to increase the production of the fibrillogenic peptide Abeta(1-42). An exception is the PSEN-1 [E318G] mutation that does not alter Abeta(1-42) generation and is generally considered a non-pathogenic polymorphism. Nevertheless, this mutation was reported to be a genetic risk factor for familial Alzheimer's disease (FAD) in the Australian population. To independently confirm this indication, we performed a case-control association study in the Italian population. We found a significant association (p<0.05, Fisher's exact test) between the presence of PSEN-1 [E318G] and FAD. In addition, on measuring the Abeta(1-42) and Abeta(1-40) concentrations in fibroblast-conditioned media cultured from PSEN-1 [E318G] carriers and PSEN-1 [wild type] controls we noted a significant decrease (p<0.05, Mann-Whitney test) in the Abeta(1-42)/Abeta(1-40) ratio in PSEN-1 [E318G] carriers, suggesting a peculiar biochemical effect of this mutation.


Subject(s)
Alzheimer Disease/genetics , Mutation , Presenilin-1/genetics , Aged , Aged, 80 and over , Amino Acid Substitution/genetics , Case-Control Studies , Female , Genetic Carrier Screening , Glutamic Acid/genetics , Glycine/genetics , Humans , Italy , Male , Middle Aged , Pedigree
SELECTION OF CITATIONS
SEARCH DETAIL
...