Gianluigi Forloni

@marionegri.it

Head of Neuroscience Department
Istituto di Ricerche Farmacologiche Mario Negri IRCCS

Dr. Forloni has served as a member of several committees of the European Community for the examination of the projects in the Neuroscience field. He is President of the Italian Association on Brain Aging Research and member of the European Academy of Sciences. He has been in charge of an elective course on genetic of neurodegenerative disorders at the Medical School of the University of Milan, he was invited for lectures and seminars in numerous Universities and Research Centers. Dr. Forloni is the author of more than 325 peer-review scientific (H index Google = 68) articles and about 30 reviews or book chapters.

EDUCATION

1976 Diploma in Industrial Chemistry, Rho (Mi)
1985 Degree in Biological Sciences, University of Milan

RESEARCH INTERESTS

Biological and genetic bases of Alzheimer’s disease (AD), Prion-related encephalopathies (PRE) and Parkinson's disease (PD). Studies on pathogenesis and possible therapeutic approaches based on the role of protein aggregation in the neurodegenerative diseases and mechanisms of neuronal death using in vitro and in vivo models

400

Scopus Publications

Scopus Publications

  • Basic Science and Pathogenesis
    Gianluigi Forloni, Claudia Balducci, Franca Orsini, Milica Cerovic, Antonio Masone, Ilaria Raimondi, Giada Lavigna, Chiara Zucchelli, Marten Beeg, Laura Colombo,et al.

    Wiley
    AbstractBackgroundThe role of oligomeric forms of various proteins as direct responsible of neuronal dysfunction in neurodegenerative disorders has been supported by numerous findings at experimental level and, more recently, by histological examinations in human material. The cellular prion protein (PrPC) has been proposed to mediate the neurotoxicity of β‐amyloid, α‐synuclein and tau oligomers. We demonstrated that although amyloid‐β oligomers (AβOs) bind with high affinity to PrPC, the memory deficit induced by intracerebroventricular (ICV) administration of AβOs in mice was not mediated by PrPC. Moreover, we did not confirm the reported interaction between α‐synuclein oligomers and PrPC, and found that their effect on memory did not depend on PrPC expression. In this study, we examined the interactions between tau oligomers (TauOs) and PrPC by chemico‐physical and functional studies.MethodTauOs were prepared in the presence of arachidonic acid and analyzed by atomic force microscopy; the interaction between TauOs and PrPC was investigated by surface plasmon resonance (SPR); the cellular localization of TauOs was studied in HEK293 cells expressing different levels of PrPC; the effect of TauOs on memory function was assessed by the novel object recognition test (NORT) after ICV injection (1µM 7.5 µl) in wild type (wt) and PrP knockout (KO) mice; the effect of TauOs on long‐term potentiation (LTP) was analyzed in hippocampal slices.ResultSPR demonstrated a high‐affinity binding between TauOs and PrPC with a Kd in the range of 20‐50 nM. Immunofluoresce analysis showed a PrPC dose‐dependent association of TauOs with the plasma membrane of HEK293 cells, and their co‐localization with PrPC. ICV application of TauOs induced memory impairment in wt but not in PrP KO mice. Accordingly, TauOs inhibited hippocampal LTP in a PrPC‐dependent fashion.ConclusionIn contrast to our previous findings with Aβ and α‐synuclein oligomers, we demonstrate that PrPC interacts with TauOs and this binding has functional consequences. This interaction might represent an interesting therapeutic target in Alzheimer’s disease and other tauopathies.

  • Creutzfeldt–Jakob disease and other prion diseases
    Inga Zerr, Anna Ladogana, Simon Mead, Peter Hermann, Gianluigi Forloni, and Brian S. Appleby

    Springer Science and Business Media LLC

  • Author Correction: Differential diagnosis of neurodegenerative dementias with the explainable MRI based machine learning algorithm MUQUBIA (Scientific Reports, (2023), 13, 1, (17355), 10.1038/s41598-023-43706-6)
    Silvia De Francesco, Claudio Crema, Damiano Archetti, Cristina Muscio, Robert I. Reid, Anna Nigri, Maria Grazia Bruzzone, Fabrizio Tagliavini, Raffaele Lodi, Egidio D’Angelo,et al.

    Springer Science and Business Media LLC

  • Evolution of brain injury and neurological dysfunction after cardiac arrest in the rat – A multimodal and comprehensive model
    Carlo Perego, Francesca Fumagalli, Francesca Motta, Marianna Cerrato, Edoardo Micotti, Davide Olivari, Daria De Giorgio, Giulia Merigo, Angelo Di Clemente, Alessandra Mandelli,et al.

    SAGE Publications
    Cardiac arrest (CA) is one of the leading causes of death worldwide. Due to hypoxic ischemic brain injury, CA survivors may experience variable degrees of neurological dysfunction. This study, for the first time, describes the progression of CA-induced neuropathology in the rat. CA rats displayed neurological and exploratory deficits. Brain MRI revealed cortical and striatal edema at 3 days (d), white matter (WM) damage in corpus callosum (CC), external capsule (EC), internal capsule (IC) at d7 and d14. At d3 a brain edema significantly correlated with neurological score. Parallel neuropathological studies showed neurodegeneration, reduced neuronal density in CA1 and hilus of hippocampus at d7 and d14, with cells dying at d3 in hilus. Microgliosis increased in cortex (Cx), caudate putamen (Cpu), CA1, CC, and EC up to d14. Astrogliosis increased earlier (d3 to d7) in Cx, Cpu, CC and EC compared to CA1 (d7 to d14). Plasma levels of neurofilament light (NfL) increased at d3 and remained elevated up to d14. NfL levels at d7 correlated with WM damage. The study shows the consequences up to 14d after CA in rats, introducing clinically relevant parameters such as advanced neuroimaging and blood biomarker useful to test therapeutic interventions in this model.

  • Transcriptomic and metabolomic changes might predict frailty in SAMP8 mice
    Letizia Dacomo, Pietro La Vitola, Laura Brunelli, Letizia Messa, Edoardo Micotti, Luisa Artioli, Elena Sinopoli, Greta Cecutti, Susanna Leva, Stella Gagliardi,et al.

    Wiley
    AbstractFrailty is a geriatric, multi‐dimensional syndrome that reflects multisystem physiological change and is a transversal measure of reduced resilience to negative events. It is characterized by weakness, frequent falls, cognitive decline, increased hospitalization and dead and represents a risk factor for the development of Alzheimer's disease (AD). The fact that frailty is recognized as a reversible condition encourages the identification of earlier biomarkers to timely predict and prevent its occurrence. SAMP8 (Senescence‐Accelerated Mouse Prone‐8) mice represent the most appropriate preclinical model to this aim and were used in this study to carry transcriptional and metabolic analyses in the brain and plasma, respectively, upon a characterization at cognitive, motor, structural, and neuropathological level at 2.5, 6, and 9 months of age. At 2.5 months, SAMP8 mice started displaying memory deficits, muscle weakness, and motor impairment. Functional alterations were associated with a neurodevelopmental deficiency associated with reduced neuronal density and glial cell loss. Through transcriptomics, we identified specific genetic signatures well distinguishing SAMP8 mice at 6 months, whereas plasma metabolomics allowed to segregate SAMP8 mice from SAMR1 already at 2.5 months of age by detecting constitutively lower levels of acylcarnitines and lipids in SAMP8 at all ages investigated correlating with functional deficits and neuropathological signs. Our findings suggest that specific genetic alterations at central level, as well as metabolomic changes in plasma, might allow to early assess a frail condition leading to dementia development, which paves the foundation for future investigation in a clinical setting.

  • Neonicotinoid Pesticides Affect Developing Neurons in Experimental Mouse Models and in Human Induced Pluripotent Stem Cell (iPSC)-Derived Neural Cultures and Organoids
    Alessandro Mariani, Davide Comolli, Roberto Fanelli, Gianluigi Forloni, and Massimiliano De Paola

    MDPI AG
    Neonicotinoids are synthetic, nicotine-derived insecticides used worldwide to protect crops and domestic animals from pest insects. The reported evidence shows that they are also able to interact with mammalian nicotine receptors (nAChRs), triggering detrimental responses in cultured neurons. Exposure to high neonicotinoid levels during the fetal period induces neurotoxicity in animal models. Considering the persistent exposure to these insecticides and the key role of nAChRs in brain development, their potential neurotoxicity on mammal central nervous system (CNS) needs further investigations. We studied here the neurodevelopmental effects of different generations of neonicotinoids on CNS cells in mouse fetal brain and primary cultures and in neuronal cells and organoids obtained from human induced pluripotent stem cells (iPSC). Neonicotinoids significantly affect neuron viability, with imidacloprid (IMI) inducing relevant alterations in synaptic protein expression, neurofilament structures, and microglia activation in vitro, and in the brain of prenatally exposed mouse fetuses. IMI induces neurotoxic effects also on developing human iPSC-derived neurons and cortical organoids. Collectively, the current findings show that neonicotinoids might induce impairment during neuro/immune-development in mouse and human CNS cells and provide new insights in the characterization of risk for the exposure to this class of pesticides.

  • Genome-wide analyses reveal a potential role for the MAPT, MOBP, and APOE loci in sporadic frontotemporal dementia
    Claudia Manzoni, Demis A. Kia, Raffaele Ferrari, Ganna Leonenko, Beatrice Costa, Valentina Saba, Edwin Jabbari, Manuela MX. Tan, Diego Albani, Victoria Alvarez,et al.

    Elsevier BV

  • SARS-CoV-2 Nucleocapsid Protein Induces Tau Pathological Changes That Can Be Counteracted by SUMO2
    Franca Orsini, Marco Bosica, Annacarla Martucci, Massimiliano De Paola, Davide Comolli, Rosaria Pascente, Gianluigi Forloni, Paul E. Fraser, Ottavio Arancio, and Luana Fioriti

    MDPI AG
    Neurologic manifestations are an immediate consequence of SARS-CoV-2 infection, the etiologic agent of COVID-19, which, however, may also trigger long-term neurological effects. Notably, COVID-19 patients with neurological symptoms show elevated levels of biomarkers associated with brain injury, including Tau proteins linked to Alzheimer’s pathology. Studies in brain organoids revealed that SARS-CoV-2 alters the phosphorylation and distribution of Tau in infected neurons, but the mechanisms are currently unknown. We hypothesize that these pathological changes are due to the recruitment of Tau into stress granules (SGs) operated by the nucleocapsid protein (NCAP) of SARS-CoV-2. To test this hypothesis, we investigated whether NCAP interacts with Tau and localizes to SGs in hippocampal neurons in vitro and in vivo. Mechanistically, we tested whether SUMOylation, a posttranslational modification of NCAP and Tau, modulates their distribution in SGs and their pathological interaction. We found that NCAP and Tau colocalize and physically interact. We also found that NCAP induces hyperphosphorylation of Tau and causes cognitive impairment in mice infected with NCAP in their hippocampus. Finally, we found that SUMOylation modulates NCAP SG formation in vitro and cognitive performance in infected mice. Our data demonstrate that NCAP induces Tau pathological changes both in vitro and in vivo. Moreover, we demonstrate that SUMO2 ameliorates NCAP-induced Tau pathology, highlighting the importance of the SUMOylation pathway as a target of intervention against neurotoxic insults, such as Tau oligomers and viral infection.

  • Genetic deletion of astrocytic calcineurin B1 prevents cognitive impairment and neuropathology development in acute and chronic mouse models of Alzheimer's disease
    Laura Tapella, Giulia Dematteis, Pietro La Vitola, Susanna Leva, Elisa Tonelli, Marco Raddi, Marta Delconti, Letizia Dacomo, Alberto La Macchia, Elisa Murari,et al.

    Wiley
    AbstractAlzheimer's disease (AD) represents an urgent yet unmet challenge for modern society, calling for exploration of innovative targets and therapeutic approaches. Astrocytes, main homeostatic cells in the CNS, represent promising cell‐target. Our aim was to investigate if deletion of the regulatory CaNB1 subunit of calcineurin in astrocytes could mitigate AD‐related memory deficits, neuropathology, and neuroinflammation. We have generated two, acute and chronic, AD mouse models with astrocytic CaNB1 ablation (ACN‐KO). In the former, we evaluated the ability of β‐amyloid oligomers (AβOs) to impair memory and activate glial cells once injected in the cerebral ventricle of conditional ACN‐KO mice. Next, we generated a tamoxifen‐inducible astrocyte‐specific CaNB1 knock‐out in 3xTg‐AD mice (indACNKO‐AD). CaNB1 was deleted, by tamoxifen injection, in 11.7‐month‐old 3xTg‐AD mice for 4.4 months. Spatial memory was evaluated using the Barnes maze; β‐amyloid plaques burden, neurofibrillary tangle deposition, reactive gliosis, and neuroinflammation were also assessed. The acute model showed that ICV injected AβOs in 2‐month‐old wild type mice impaired recognition memory and fostered a pro‐inflammatory microglia phenotype, whereas in ACN‐KO mice, AβOs were inactive. In indACNKO‐AD mice, 4.4 months after CaNB1 depletion, we found preservation of spatial memory and cognitive flexibility, abolishment of amyloidosis, and reduction of neurofibrillary tangles, gliosis, and neuroinflammation. Our results suggest that ACN is crucial for the development of cognitive impairment, AD neuropathology, and neuroinflammation. Astrocyte‐specific CaNB1 deletion is beneficial for both the abolishment of AβO‐mediated detrimental effects and treatment of ongoing AD‐related pathology, hence representing an intriguing target for AD therapy.

  • Synergistic Pharmacological Therapy to Modulate Glial Cells in Spinal Cord Injury
    Valeria Veneruso, Emilia Petillo, Fabio Pizzetti, Alessandro Orro, Davide Comolli, Massimiliano De Paola, Antonietta Verrillo, Arianna Baggiolini, Simona Votano, Franca Castiglione,et al.

    Wiley
    AbstractCurrent treatments for modulating the glial‐mediated inflammatory response after spinal cord injury (SCI) have limited ability to improve recovery. This is quite likely due to the lack of a selective therapeutic approach acting on microgliosis and astrocytosis, the glia components most involved after trauma, while maximizing efficacy and minimizing side effects. A new nanogel that can selectively release active compounds in microglial cells and astrocytes is developed and characterized. The degree of selectivity and subcellular distribution of the nanogel is evaluated by applying an innovative super‐resolution microscopy technique, expansion microscopy. Two different administration schemes are then tested in a SCI mouse model: in an early phase, the nanogel loaded with Rolipram, an anti‐inflammatory drug, achieves significant improvement in the animal's motor performance due to the increased recruitment of microglia and macrophages that are able to localize the lesion. Treatment in the late phase, however, gives opposite results, with worse motor recovery because of the widespread degeneration. These findings demonstrate that the nanovector can be selective and functional in the treatment of the glial component in different phases of SCI. They also open a new therapeutic scenario for tackling glia‐mediated inflammation after neurodegenerative events in the central nervous system.

  • Polygenic scores of subcortical brain volumes as possible modulators of treatment response in depression
    Vincenzo Oliva, Alfonso Martone, Giuseppe Fanelli, Katharina Domschke, Alessandra Minelli, Massimo Gennarelli, Paolo Martini, Marco Bortolomasi, Eduard Maron, Alessio Squassina,et al.

    Elsevier BV

  • Differential diagnosis of neurodegenerative dementias with the explainable MRI based machine learning algorithm MUQUBIA
    Silvia De Francesco, Claudio Crema, Damiano Archetti, Cristina Muscio, Robert I. Reid, Anna Nigri, Maria Grazia Bruzzone, Fabrizio Tagliavini, Raffaele Lodi, Egidio D’Angelo,et al.

    Springer Science and Business Media LLC
    AbstractBiomarker-based differential diagnosis of the most common forms of dementia is becoming increasingly important. Machine learning (ML) may be able to address this challenge. The aim of this study was to develop and interpret a ML algorithm capable of differentiating Alzheimer’s dementia, frontotemporal dementia, dementia with Lewy bodies and cognitively normal control subjects based on sociodemographic, clinical, and magnetic resonance imaging (MRI) variables. 506 subjects from 5 databases were included. MRI images were processed with FreeSurfer, LPA, and TRACULA to obtain brain volumes and thicknesses, white matter lesions and diffusion metrics. MRI metrics were used in conjunction with clinical and demographic data to perform differential diagnosis based on a Support Vector Machine model called MUQUBIA (Multimodal Quantification of Brain whIte matter biomArkers). Age, gender, Clinical Dementia Rating (CDR) Dementia Staging Instrument, and 19 imaging features formed the best set of discriminative features. The predictive model performed with an overall Area Under the Curve of 98%, high overall precision (88%), recall (88%), and F1 scores (88%) in the test group, and good Label Ranking Average Precision score (0.95) in a subset of neuropathologically assessed patients. The results of MUQUBIA were explained by the SHapley Additive exPlanations (SHAP) method. The MUQUBIA algorithm successfully classified various dementias with good performance using cost-effective clinical and MRI information, and with independent validation, has the potential to assist physicians in their clinical diagnosis.

  • Oligomers and Neurodegeneration: New Evidence
    Gianluigi Forloni

    Aging and Disease
    In the last few months new results in Alzheimer’s (AD) and Parkinson’s disease (PD) have converged, attracting attention to oligomer species of misfolded proteins, β-amyloid (Aβ and α-synuclein (α-Syn), in the pathogenesis. The high affinity for Aβ protofibrils and oligomers of lecanemab, an antibody recently approved as a disease-modifying drug in AD, and the identification of Aβ-oligomers in blood samples as early biomarkers in subjects with cognitive decline, indicate the oligomers as a therapeutic target and diagnostic tool in AD. α-Syn oligomers were identified by new histopathological techniques in the hippocampus and visual cortex of PD subjects with a distribution distinct from the Lewy body pathologies but associated with cognitive impairment; these species purified from PD brain were highly neurotoxic. In a PD experimental model, we confirmed the presence of α-Syn oligomers associated with cognitive decline and sensitive to drug treatment.

  • Development of an Induced Pluripotent Stem Cell-Based Liver-on-a-Chip Assessed with an Alzheimer's Disease Drug
    Francesca Fanizza, Lucia Boeri, Francesca Donnaloja, Simone Perottoni, Gianluigi Forloni, Carmen Giordano, and Diego Albani

    American Chemical Society (ACS)
    Liver-related drug metabolism is a key aspect of pharmacokinetics and possible toxicity. From this perspective, the availability of advanced in vitro models for drug testing is still an open need, also to the end of reducing the burden of in vivo experiments. In this scenario, organ-on-a-chip is gaining attention as it couples a state-of-the art in vitro approach to the recapitulation of key in vivo physiological features such as fluidodynamics and a tri-dimensional cytoarchitecture. We implemented a novel liver-on-a-chip (LoC) device based on an innovative dynamic device (MINERVA 2.0) where functional hepatocytes (iHep) have been encapsulated into a 3D hydrogel matrix interfaced through a porous membrane with endothelial cells (iEndo)]. Both lines were derived from human-induced pluripotent stem cells (iPSCs), and the LoC was functionally assessed with donepezil, a drug approved for Alzheimer’s disease therapy. The presence of iEndo and a 3D microenvironment enhanced the expression of liver-specific physiologic functions as in iHep, after 7 day perfusion, we noticed an increase of albumin, urea production, and cytochrome CYP3A4 expression compared to the iHep static culture. In particular, for donepezil kinetics, a computational fluid dynamic study conducted to assess the amount of donepezil diffused into the LoC indicated that the molecule should be able to pass through the iEndo and reach the target iHep construct. Then, we performed experiments of donepezil kinetics that confirmed the numerical simulations. Overall, our iPSC-based LoC reproduced the in vivo physiological microenvironment of the liver and was suitable for potential hepatotoxic screening studies.

  • Author Correction: A consensus protocol for functional connectivity analysis in the rat brain (Nature Neuroscience, (2023), 26, 4, (673-681), 10.1038/s41593-023-01286-8)
    Joanes Grandjean, Gabriel Desrosiers-Gregoire, Cynthia Anckaerts, Diego Angeles-Valdez, Fadi Ayad, David A. Barrière, Ines Blockx, Aleksandra Bortel, Margaret Broadwater, Beatriz M. Cardoso,et al.

    Springer Science and Business Media LLC

  • Prioritization of Drug Targets for Neurodegenerative Diseases by Integrating Genetic and Proteomic Data From Brain and Blood
    Yi-Jun Ge, Ya-Nan Ou, Yue-Ting Deng, Bang-Sheng Wu, Liu Yang, Ya-Ru Zhang, Shi-Dong Chen, Yu-Yuan Huang, Qiang Dong, Lan Tan,et al.

    Elsevier BV

  • Neural cortical organoids from self-assembling human iPSC as a model to investigate neurotoxicity in brain ischemia
    Massimiliano De Paola, Francesca Pischiutta, Davide Comolli, Alessandro Mariani, Joe Kelk, Ilaria Lisi, Milica Cerovic, Stefano Fumagalli, Gianluigi Forloni, and Elisa R Zanier

    SAGE Publications
    Brain ischemia is a common acute injury resulting from impaired blood flow to the brain. Translation of effective drug candidates from experimental models to patients has systematically failed. The use of human induced pluripotent stem cells (iPSC) offers new opportunities to gain translational insights into diseases including brain ischemia. We used a human 3D self-assembling iPSC-derived model (human cortical organoids, hCO) to characterize the effects of ischemia caused by oxygen-glucose deprivation (OGD). hCO exposed to 2 h or 8 h of OGD had neuronal death and impaired neuronal network complexity, measured in whole-mounting microtubule-associated protein 2 (MAP-2) immunostaining. Neuronal vulnerability was reflected by a reduction in MAP-2 mRNA levels, and increased release of neurofilament light chain (NfL) in culture media, proportional to OGD severity. Glial fibrillary acidic protein (GFAP) gene or protein levels did not change in hCO, but their release in medium increased after prolonged OGD. In conclusion, this human 3D iPSC-based in vitro model of brain ischemic injury is characterized by marked neuronal injury reflected by the release of the translational biomarker NfL which is relevant for testing neuroprotective strategies.

  • A consensus protocol for functional connectivity analysis in the rat brain
    Joanes Grandjean, Gabriel Desrosiers-Gregoire, Cynthia Anckaerts, Diego Angeles-Valdez, Fadi Ayad, David A. Barrière, Ines Blockx, Aleksandra Bortel, Margaret Broadwater, Beatriz M. Cardoso,et al.

    Springer Science and Business Media LLC

  • Alpha Synuclein: Neurodegeneration and Inflammation
    Gianluigi Forloni

    MDPI AG
    Alpha-Synuclein (α-Syn) is one of the most important molecules involved in the pathogenesis of Parkinson’s disease and related disorders, synucleinopathies, but also in several other neurodegenerative disorders with a more elusive role. This review analyzes the activities of α-Syn, in different conformational states, monomeric, oligomeric and fibrils, in relation to neuronal dysfunction. The neuronal damage induced by α-Syn in various conformers will be analyzed in relation to its capacity to spread the intracellular aggregation seeds with a prion-like mechanism. In view of the prominent role of inflammation in virtually all neurodegenerative disorders, the activity of α-Syn will also be illustrated considering its influence on glial reactivity. We and others have described the interaction between general inflammation and cerebral dysfunctional activity of α-Syn. Differences in microglia and astrocyte activation have also been observed when in vivo the presence of α-Syn oligomers has been combined with a lasting peripheral inflammatory effect. The reactivity of microglia was amplified, while astrocytes were damaged by the double stimulus, opening new perspectives for the control of inflammation in synucleinopathies. Starting from our studies in experimental models, we extended the perspective to find useful pointers to orient future research and potential therapeutic strategies in neurodegenerative disorders.

  • Repositioning doxycycline for treating synucleinopathies: Evidence from a pre-clinical mouse model
    Pietro La Vitola, Luisa Artioli, Milica Cerovic, Cristian Poletto, Letizia Dacomo, Susanna Leva, Claudia Balducci, and Gianluigi Forloni

    Elsevier BV

  • Melancholic features and typical neurovegetative symptoms of major depressive disorder show specific polygenic patterns
    Vincenzo Oliva, Giuseppe Fanelli, Siegfried Kasper, Joseph Zohar, Daniel Souery, Stuart Montgomery, Diego Albani, Gianluigi Forloni, Panagiotis Ferentinos, Dan Rujescu,et al.

    Elsevier BV

  • MRI data quality assessment for the RIN - Neuroimaging Network using the ACR phantoms
    Fulvia Palesi, Anna Nigri, Ruben Gianeri, Domenico Aquino, Alberto Redolfi, Laura Biagi, Irene Carne, Silvia De Francesco, Stefania Ferraro, Paola Martucci,et al.

    Elsevier BV

  • Multi-centre and multi-vendor reproducibility of a standardized protocol for quantitative susceptibility Mapping of the human brain at 3T
    Marta Lancione, Paolo Bosco, Mauro Costagli, Anna Nigri, Domenico Aquino, Irene Carne, Stefania Ferraro, Giovanni Giulietti, Antonio Napolitano, Fulvia Palesi,et al.

    Elsevier BV

  • Oligomeropathies, inflammation and prion protein binding
    Gianluigi Forloni, Pietro La Vitola, and Claudia Balducci

    Frontiers Media SA
    The central role of oligomers, small soluble aggregates of misfolded proteins, in the pathogenesis of neurodegenerative disorders is recognized in numerous experimental conditions and is compatible with clinical evidence. To underline this concept, some years ago we coined the term oligomeropathies to define the common mechanism of action of protein misfolding diseases like Alzheimer, Parkinson or prion diseases. Using simple experimental conditions, with direct application of synthetic β amyloid or α-synuclein oligomers intraventricularly at micromolar concentrations, we could detect differences and similarities in the biological consequences. The two oligomer species affected cognitive behavior, neuronal dysfunction and cerebral inflammatory reactions with distinct mechanisms. In these experimental conditions the proposed mediatory role of cellular prion protein in oligomer activities was not confirmed. Together with oligomers, inflammation at different levels can be important early in neurodegenerative disorders; both β amyloid and α-synuclein oligomers induce inflammation and its control strongly affects neuronal dysfunction. This review summarizes our studies with β-amyloid or α-synuclein oligomers, also considering the potential curative role of doxycycline, a well-known antibiotic with anti-amyloidogenic and anti-inflammatory activities. These actions are analyzed in terms of the therapeutic prospects.

  • The development of ADAM10 endocytosis inhibitors for the treatment of Alzheimer's disease
    Stefano Musardo, Sebastien Therin, Silvia Pelucchi, Laura D’Andrea, Ramona Stringhi, Ana Ribeiro, Annalisa Manca, Claudia Balducci, Jessica Pagano, Carlo Sala,et al.

    Elsevier BV