85 results on '"Monica Nizzardo"'
Search Results
2. Systematic elucidation of neuron-astrocyte interaction in models of amyotrophic lateral sclerosis using multi-modal integrated bioinformatics workflow
- Author
-
Vartika Mishra, Diane B. Re, Virginia Le Verche, Mariano J. Alvarez, Alessandro Vasciaveo, Arnaud Jacquier, Paschalis-Tomas Doulias, Todd M. Greco, Monica Nizzardo, Dimitra Papadimitriou, Tetsuya Nagata, Paola Rinchetti, Eduardo J. Perez-Torres, Kristin A. Politi, Burcin Ikiz, Kevin Clare, Manuel E. Than, Stefania Corti, Harry Ischiropoulos, Francesco Lotti, Andrea Califano, and Serge Przedborski
- Subjects
Science - Abstract
Neuron-astrocyte communication plays a key role in pathophysiology, however systematic approaches to unveil it are limited. Here, the authors propose SEARCHIN, a multi-modal integrated workflow, as a tool to identify cross-compartment ligand-receptor interactions, applied to ALS models.
- Published
- 2020
- Full Text
- View/download PDF
3. TDP-43 promotes the formation of neuromuscular synapses through the regulation of Disc-large expression in Drosophila skeletal muscles
- Author
-
Nina Strah, Giulia Romano, Clelia Introna, Raffaella Klima, Marta Marzullo, Laura Ciapponi, Aram Megighian, Monica Nizzardo, and Fabian Feiguin
- Subjects
TDP-43 ,Skeletal muscles ,Dlg ,Neuromuscular junctions ,ALS ,Biology (General) ,QH301-705.5 - Abstract
Abstract Background The ribonuclear protein TDP-43 has been implicated in the pathophysiology of amyotrophic lateral sclerosis (ALS), with genetic mutations being linked to the neurological symptoms of the disease. Though alterations in the intracellular distribution of TDP-43 have been observed in skeletal muscles of patients suffering from ALS, it is not clear whether such modifications play an active role in the disease or merely represent an expression of muscle homeostatic mechanisms. Also, the molecular and metabolic pathways regulated by TDP-43 in the skeletal muscle remain largely unknown. Here, we analyze the function of TBPH, the Drosophila melanogaster ortholog of TDP-43, in skeletal muscles. Results We modulated the activity of TDP-43 in Drosophila muscles by means of RNA interference and observed that it is required to promote the formation and growth of neuromuscular synapses. TDP-43 regulated the expression levels of Disc-large (Dlg), and restoring Dlg expression either in skeletal muscles or in motoneurons was sufficient to suppress the locomotive and synaptic defects of TDP-43-null flies. These results were validated by the observation of a decrease in Dlg levels in human neuroblastoma cells and iPSC-differentiated motoneurons derived from ALS patients, suggesting similar mechanisms may potentially be involved in the pathophysiology of the disease. Conclusions Our results help to unveil the physiological role of TDP-43 in skeletal muscles as well as the mechanisms responsible for the autonomous and non-autonomous behavior of this protein concerning the organization of neuromuscular synapses.
- Published
- 2020
- Full Text
- View/download PDF
4. Stathmins and Motor Neuron Diseases: Pathophysiology and Therapeutic Targets
- Author
-
Delia Gagliardi, Elisa Pagliari, Megi Meneri, Valentina Melzi, Federica Rizzo, Giacomo Pietro Comi, Stefania Corti, Michela Taiana, and Monica Nizzardo
- Subjects
stathmin ,motor neuron diseases ,ALS ,SMA ,STMN2 ,STMN1 ,Biology (General) ,QH301-705.5 - Abstract
Motor neuron diseases (MNDs) are a group of fatal, neurodegenerative disorders with different etiology, clinical course and presentation, caused by the loss of upper and lower motor neurons (MNs). MNs are highly specialized cells equipped with long, axonal processes; axonal defects are some of the main players underlying the pathogenesis of these disorders. Microtubules are key components of the neuronal cytoskeleton characterized by dynamic instability, switching between rapid polymerization and shrinkage. Proteins of the stathmin family affect microtubule dynamics regulating the assembly and the dismantling of tubulin. Stathmin-2 (STMN2) is one of the most abundantly expressed genes in MNs. Following axonal injury, STMN2 expression is upregulated, and the protein is transported toward the growth cones of regenerating axons. STMN2 has a critical role in axonal maintenance, and its dysregulation plays an important role in neurodegenerative processes. Stathmin-1 (STMN1) is a ubiquitous protein that is highly expressed during the development of the nervous system, and its phosphorylation controls microtubule dynamics. In the present review, we summarize what is currently known about the involvement of stathmin alterations in MNDs and the potential therapeutic effect of their modulation, with a specific focus on the most common forms of MND, amyotrophic lateral sclerosis (ALS) and spinal muscular atrophy (SMA).
- Published
- 2022
- Full Text
- View/download PDF
5. Targeting PTB for Glia-to-Neuron Reprogramming In Vitro and In Vivo for Therapeutic Development in Neurological Diseases
- Author
-
Matilde Contardo, Roberta De Gioia, Delia Gagliardi, Giacomo Pietro Comi, Linda Ottoboni, Monica Nizzardo, and Stefania Corti
- Subjects
PTB ,reprogramming ,neuron ,neurodegenerative diseases ,Biology (General) ,QH301-705.5 - Abstract
In vivo cell reprogramming of glial cells offers a promising way to generate new neurons in the adult mammalian nervous system. This approach might compensate for neuronal loss occurring in neurological disorders, but clinically viable tools are needed to advance this strategy from bench to bedside. Recently published work has described the successful neuronal conversion of glial cells through the repression of a single gene, polypyrimidine tract-binding protein 1 (Ptbp1), which encodes a key RNA-binding protein. Newly converted neurons not only express correct markers but they also functionally integrate into endogenous brain circuits and modify disease symptoms in in vivo models of neurodegenerative diseases. However, doubts about the nature of “converted” neurons, in particular in vivo, have been raised, based on concerns about tracking reporter genes in converted cells. More robust lineage tracing is needed to draw definitive conclusions about the reliability of this strategy. In vivo reprogramming and the possibility of implementing it with approaches that could be translated into the clinic with antisense oligonucleotides targeting a single gene like Ptbp1 are hot topics. They warrant further investigation with stringent methods and criteria of evaluation for the ultimate treatment of neurological diseases.
- Published
- 2022
- Full Text
- View/download PDF
6. Mitochondrial Dysregulation and Impaired Autophagy in iPSC-Derived Dopaminergic Neurons of Multiple System Atrophy
- Author
-
Giacomo Monzio Compagnoni, Giulio Kleiner, Maura Samarani, Massimo Aureli, Gaia Faustini, Arianna Bellucci, Dario Ronchi, Andreina Bordoni, Manuela Garbellini, Sabrina Salani, Francesco Fortunato, Emanuele Frattini, Elena Abati, Christian Bergamini, Romana Fato, Silvia Tabano, Monica Miozzo, Giulia Serratto, Maria Passafaro, Michela Deleidi, Rosamaria Silipigni, Monica Nizzardo, Nereo Bresolin, Giacomo P. Comi, Stefania Corti, Catarina M. Quinzii, and Alessio Di Fonzo
- Subjects
Medicine (General) ,R5-920 ,Biology (General) ,QH301-705.5 - Abstract
Summary: Multiple system atrophy (MSA) is a progressive neurodegenerative disease that affects several areas of the CNS, whose pathogenesis is still widely unclear and for which an effective treatment is lacking. We have generated induced pluripotent stem cell-derived dopaminergic neurons from four MSA patients and four healthy controls and from two monozygotic twins discordant for the disease. In this model, we have demonstrated an aberrant autophagic flow and a mitochondrial dysregulation involving respiratory chain activity, mitochondrial content, and CoQ10 biosynthesis. These defective mechanisms may contribute to the onset of the disease, representing potential therapeutic targets. : Monzio Compagnoni et al. present an iPSC-based neuronal in vitro model of multiple system atrophy. Patients' dopaminergic neurons display a dysregulation of mitochondrial functioning and autophagy, suggesting new hints for the comprehension of the pathogenesis of the disease. Keywords: multiple system atrophy, induced pluripotent stem cells, dopaminergic neurons, mitochondria, autophagy, MSA, neurodegeneration
- Published
- 2018
- Full Text
- View/download PDF
7. Dysregulation of Muscle-Specific MicroRNAs as Common Pathogenic Feature Associated with Muscle Atrophy in ALS, SMA and SBMA: Evidence from Animal Models and Human Patients
- Author
-
Claudia Malacarne, Mariarita Galbiati, Eleonora Giagnorio, Paola Cavalcante, Franco Salerno, Francesca Andreetta, Cinza Cagnoli, Michela Taiana, Monica Nizzardo, Stefania Corti, Viviana Pensato, Anna Venerando, Cinzia Gellera, Silvia Fenu, Davide Pareyson, Riccardo Masson, Lorenzo Maggi, Eleonora Dalla Bella, Giuseppe Lauria, Renato Mantegazza, Pia Bernasconi, Angelo Poletti, Silvia Bonanno, and Stefania Marcuzzo
- Subjects
motor neuron diseases ,muscle-specific microRNAs ,amyotrophic lateral sclerosis ,spinal muscular atrophy ,spinal bulbar muscular atrophy ,mouse models ,Biology (General) ,QH301-705.5 ,Chemistry ,QD1-999 - Abstract
Motor neuron diseases (MNDs) are neurodegenerative disorders characterized by upper and/or lower MN loss. MNDs include amyotrophic lateral sclerosis (ALS), spinal muscular atrophy (SMA), and spinal and bulbar muscular atrophy (SBMA). Despite variability in onset, progression, and genetics, they share a common skeletal muscle involvement, suggesting that it could be a primary site for MND pathogenesis. Due to the key role of muscle-specific microRNAs (myomiRs) in skeletal muscle development, by real-time PCR we investigated the expression of miR-206, miR-133a, miR-133b, and miR-1, and their target genes, in G93A-SOD1 ALS, Δ7SMA, and KI-SBMA mouse muscle during disease progression. Further, we analyzed their expression in serum of SOD1-mutated ALS, SMA, and SBMA patients, to demonstrate myomiR role as noninvasive biomarkers. Our data showed a dysregulation of myomiRs and their targets, in ALS, SMA, and SBMA mice, revealing a common pathogenic feature associated with muscle impairment. A similar myomiR signature was observed in patients’ sera. In particular, an up-regulation of miR-206 was identified in both mouse muscle and serum of human patients. Our overall findings highlight the role of myomiRs as promising biomarkers in ALS, SMA, and SBMA. Further investigations are needed to explore the potential of myomiRs as therapeutic targets for MND treatment.
- Published
- 2021
- Full Text
- View/download PDF
8. Neural Stem Cell Transplantation for Neurodegenerative Diseases
- Author
-
Roberta De Gioia, Fabio Biella, Gaia Citterio, Federica Rizzo, Elena Abati, Monica Nizzardo, Nereo Bresolin, Giacomo Pietro Comi, and Stefania Corti
- Subjects
neuronal stem cells ,neural subpopulation ,neurodegenerative disease ,cell therapy ,Biology (General) ,QH301-705.5 ,Chemistry ,QD1-999 - Abstract
Neurodegenerative diseases are disabling and fatal neurological disorders that currently lack effective treatment. Neural stem cell (NSC) transplantation has been studied as a potential therapeutic approach and appears to exert a beneficial effect against neurodegeneration via different mechanisms, such as the production of neurotrophic factors, decreased neuroinflammation, enhanced neuronal plasticity and cell replacement. Thus, NSC transplantation may represent an effective therapeutic strategy. To exploit NSCs’ potential, some of their essential biological characteristics must be thoroughly investigated, including the specific markers for NSC subpopulations, to allow profiling and selection. Another key feature is their secretome, which is responsible for the regulation of intercellular communication, neuroprotection, and immunomodulation. In addition, NSCs must properly migrate into the central nervous system (CNS) and integrate into host neuronal circuits, enhancing neuroplasticity. Understanding and modulating these aspects can allow us to further exploit the therapeutic potential of NSCs. Recent progress in gene editing and cellular engineering techniques has opened up the possibility of modifying NSCs to express select candidate molecules to further enhance their therapeutic effects. This review summarizes current knowledge regarding these aspects, promoting the development of stem cell therapies that could be applied safely and effectively in clinical settings.
- Published
- 2020
- Full Text
- View/download PDF
9. iPSC-Derived Neural Stem Cells Act via Kinase Inhibition to Exert Neuroprotective Effects in Spinal Muscular Atrophy with Respiratory Distress Type 1
- Author
-
Chiara Simone, Monica Nizzardo, Federica Rizzo, Margherita Ruggieri, Giulietta Riboldi, Sabrina Salani, Monica Bucchia, Nereo Bresolin, Giacomo P. Comi, and Stefania Corti
- Subjects
Medicine (General) ,R5-920 ,Biology (General) ,QH301-705.5 - Abstract
Spinal muscular atrophy with respiratory distress type 1 (SMARD1) is a motor neuron disease caused by mutations in the IGHMBP2 gene, without a cure. Here, we demonstrate that neural stem cells (NSCs) from human-induced pluripotent stem cells (iPSCs) have therapeutic potential in the context of SMARD1. We show that upon transplantation NSCs can appropriately engraft and differentiate in the spinal cord of SMARD1 animals, ameliorating their phenotype, by protecting their endogenous motor neurons. To evaluate the effect of NSCs in the context of human disease, we generated human SMARD1-iPSCs motor neurons that had a significantly reduced survival and axon length. Notably, the coculture with NSCs ameliorate these disease features, an effect attributable to the production of neurotrophic factors and their dual inhibition of GSK-3 and HGK kinases. Our data support the role of iPSC as SMARD1 disease model and their translational potential for therapies in motor neuron disorders.
- Published
- 2014
- Full Text
- View/download PDF
10. Investigation of New Morpholino Oligomers to Increase Survival Motor Neuron Protein Levels in Spinal Muscular Atrophy
- Author
-
Agnese Ramirez, Sebastiano G. Crisafulli, Mafalda Rizzuti, Nereo Bresolin, Giacomo P. Comi, Stefania Corti, and Monica Nizzardo
- Subjects
spinal muscular atrophy ,morpholino ,therapy ,Biology (General) ,QH301-705.5 ,Chemistry ,QD1-999 - Abstract
Spinal muscular atrophy (SMA) is an autosomal-recessive childhood motor neuron disease and the main genetic cause of infant mortality. SMA is caused by deletions or mutations in the survival motor neuron 1 (SMN1) gene, which results in SMN protein deficiency. Only one approved drug has recently become available and allows for the correction of aberrant splicing of the paralogous SMN2 gene by antisense oligonucleotides (ASOs), leading to production of full-length SMN protein. We have already demonstrated that a sequence of an ASO variant, Morpholino (MO), is particularly suitable because of its safety and efficacy profile and is both able to increase SMN levels and rescue the murine SMA phenotype. Here, we optimized this strategy by testing the efficacy of four new MO sequences targeting SMN2. Two out of the four new MO sequences showed better efficacy in terms of SMN protein production both in SMA induced pluripotent stem cells (iPSCs) and SMAΔ7 mice. Further, the effect was enhanced when different MO sequences were administered in combination. Our data provide an important insight for MO-based treatment for SMA. Optimization of the target sequence and validation of a treatment based on a combination of different MO sequences could support further pre-clinical studies and the progression toward future clinical trials.
- Published
- 2018
- Full Text
- View/download PDF
11. Direct Reprogramming of Adult Somatic Cells into other Lineages: Past Evidence and Future Perspectives
- Author
-
Monica Nizzardo, Chiara Simone, Marianna Falcone, Giulietta Riboldi, Giacomo P. Comi, Nereo Bresolin, and Stefania Corti
- Subjects
Medicine - Abstract
Direct reprogramming of an adult cell into another differentiated lineage—such as fibroblasts into neurons, cardiomyocytes, or blood cells—without passage through an undifferentiated pluripotent stage is a new area of research that has recently emerged alongside stem cell technology and induced pluripotent stem cell reprogramming; indeed, this avenue of investigation has begun to play a central role in basic biological research and regenerative medicine. Even though the field seems new, its origins go back to the 1980s when it was demonstrated that differentiated adult cells can be converted into another cell lineage through the overexpression of transcription factors, establishing mature cell plasticity. Here, we retrace transdifferentiation experiments from the discovery of master control genes to recent in vivo reprogramming of one somatic cell into another from the perspective of possible applications for the development of new therapeutic approaches for human diseases.
- Published
- 2013
- Full Text
- View/download PDF
12. Combined RNA interference and gene replacement therapy targeting MFN2 for the treatment of Charcot-Marie-Tooth type 2A
- Author
-
Federica Rizzo, Silvia Bono, Marc David Ruepp, Sabrina Salani, Linda Ottoboni, Elena Abati, Valentina Melzi, Chiara Cordiglieri, Serena Pagliarani, Roberta De Gioia, Alessia Anastasia, Michela Taiana, Manuela Garbellini, Simona Lodato, Paolo Kunderfranco, Daniele Cazzato, Daniele Cartelli, Caterina Lonati, Nereo Bresolin, Giacomo Comi, Monica Nizzardo, and S Corti
- Abstract
Mitofusin-2 (MFN2) is an outer mitochondrial membrane protein essential for mitochondrial networking in most cells. Autosomal dominant mutations in the MFN2 gene cause Charcot-Marie-Tooth type 2A disease (CMT2A), a severe and disabling sensory-motor neuropathy that impacts the entire nervous system. Here, we propose a novel therapeutic strategy tailored to correcting the root genetic defect of CMT2A. Though mutant and wild-type MFN2 mRNA are inhibited by RNA interference (RNAi), the wild-type protein is restored by overexpressing cDNA encoding functional MFN2 modified to be resistant to RNAi. We tested this strategy in CMT2A patient-specific human induced pluripotent stem cell (iPSC)-differentiated motor neurons (MNs), demonstrating the correct silencing of endogenous MFN2 and replacement with an exogenous copy of the functional wild-type gene. This approach significantly rescues the CMT2A MN phenotype in vitro, stabilizing the altered axonal mitochondrial distribution and correcting abnormal mitophagic processes. The MFN2molecular correction was also properly confirmed in vivo in the MitoCharc1 CMT2A transgenic mouse model after cerebrospinal fluid (CSF) delivery of the constructs into newborn mice using adeno-associated virus 9 (AAV9). Altogether, our data support the feasibility of a combined RNAi and gene therapy strategy for treating the broad spectrum of human diseases associated with MFN2mutations.
- Published
- 2023
13. Molecular analysis of SMARD1 patient-derived cells demonstrates that nonsense-mediated mRNA decay is impaired
- Author
-
Michela Taiana, Alessandra Govoni, Sabrina Salani, Nicole Kleinschmidt, Noemi Galli, Matteo Saladini, Stefano Bruno Ghezzi, Valentina Melzi, Margherita Bersani, Roberto Del Bo, Oliver Muehlemann, Enrico Bertini, Valeria Sansone, Emilio Albamonte, Sonia Messina, Francesco Mari, Elisabetta Cesaroni, Liliana Porfiri, Francesco Danilo Tiziano, Gian Luca Vita, Maria Sframeli, Carmen Bonanno, Nereo Bresolin, Giacomo Comi, Stefania Corti, and Monica Nizzardo
- Subjects
Muscular Atrophy, Spinal ,Respiratory Distress Syndrome, Newborn ,Psychiatry and Mental health ,Infant, Newborn ,motor neuron disease ,Humans ,Surgery ,Neurology (clinical) ,Settore MED/03 - GENETICA MEDICA ,Nonsense Mediated mRNA Decay - Published
- 2022
14. Insights into the identification of a molecular signature for amyotrophic lateral sclerosis exploiting integrated microRNA profiling of iPSC-derived motor neurons and exosomes
- Author
-
Mafalda Rizzuti, Valentina Melzi, Delia Gagliardi, Davide Resnati, Megi Meneri, Laura Dioni, Pegah Masrori, Nicole Hersmus, Koen Poesen, Martina Locatelli, Fabio Biella, Rosamaria Silipigni, Valentina Bollati, Nereo Bresolin, Giacomo Pietro Comi, Philip Van Damme, Monica Nizzardo, and Stefania Corti
- Subjects
EXPRESSION ,Biochemistry & Molecular Biology ,STRESS ,TDP-43 ,Induced Pluripotent Stem Cells ,BIOMARKERS ,C9ORF72 ,CSF ,Cell Communication ,Exosomes ,Cellular and Molecular Neuroscience ,Humans ,Molecular Biology ,Cells, Cultured ,miRNA ,Motor neurons ,Motor Neurons ,Pharmacology ,Science & Technology ,HEXANUCLEOTIDE REPEAT ,MIRNA BIOGENESIS ,MUTATIONS ,Amyotrophic Lateral Sclerosis ,Cell Biology ,GENE ,MicroRNAs ,DIFFERENTIATION ,Gene Expression Regulation ,Case-Control Studies ,Molecular Medicine ,ALS ,Life Sciences & Biomedicine - Abstract
Amyotrophic lateral sclerosis (ALS) is a rare neurodegenerative disorder characterized by progressive degeneration of motor neurons (MNs). Most cases are sporadic, whereas 10% are familial. The pathological mechanisms underlying the disease are partially understood, but it is increasingly being recognized that alterations in RNA metabolism and deregulation of microRNA (miRNA) expression occur in ALS. In this study, we performed miRNA expression profile analysis of iPSC-derived MNs and related exosomes from familial patients and healthy subjects. We identified dysregulation of miR-34a, miR-335 and miR-625-3p expression in both MNs and exosomes. These miRNAs regulate genes and pathways which correlate with disease pathogenesis, suggesting that studying miRNAs deregulation can contribute to deeply investigate the molecular mechanisms underlying the disease. We also assayed the expression profile of these miRNAs in the cerebrospinal fluid (CSF) of familial (fALS) and sporadic patients (sALS) and we identified a significant dysregulation of miR-34a-3p and miR-625-3p levels in ALS compared to controls. Taken together, all these findings suggest that miRNA analysis simultaneously performed in different human biological samples could represent a promising molecular tool to understand the etiopathogenesis of ALS and to develop new potential miRNA-based strategies in this new propitious therapeutic era. ispartof: CELLULAR AND MOLECULAR LIFE SCIENCES vol:79 issue:3 ispartof: location:Switzerland status: published
- Published
- 2022
15. Systematic elucidation of neuron-astrocyte interaction in models of amyotrophic lateral sclerosis using multi-modal integrated bioinformatics workflow
- Author
-
Manuel E. Than, Burcin Ikiz, Virginia Le Verche, Paola Rinchetti, Arnaud Jacquier, Vartika Mishra, Harry Ischiropoulos, Todd M. Greco, Kristin A. Politi, Dimitra Papadimitriou, Mariano J. Alvarez, Kevin Clare, Francesco Lotti, Alessandro Vasciaveo, Paschalis Tomas Doulias, Serge Przedborski, Diane B. Re, Andrea Califano, Monica Nizzardo, Stefania Corti, Eduardo J. Perez-Torres, and Tetsuya Nagata
- Subjects
0301 basic medicine ,Proteomics ,General Physics and Astronomy ,Cell Communication ,Ligands ,Receptors, Tumor Necrosis Factor ,Amyloid beta-Protein Precursor ,Mice ,0302 clinical medicine ,Superoxide Dismutase-1 ,Amyloid precursor protein ,Amyotrophic lateral sclerosis ,RNA, Small Interfering ,lcsh:Science ,Cells, Cultured ,Motor Neurons ,Multidisciplinary ,biology ,Cell Death ,Phenotype ,medicine.anatomical_structure ,Matrix Metalloproteinase 9 ,Gene Knockdown Techniques ,Systems biology ,Science ,Systems analysis ,Context (language use) ,Mice, Transgenic ,Molecular neuroscience ,General Biochemistry, Genetics and Molecular Biology ,Article ,03 medical and health sciences ,medicine ,Animals ,Humans ,Gene Silencing ,Mechanism (biology) ,Amyotrophic Lateral Sclerosis ,Computational Biology ,General Chemistry ,medicine.disease ,Disease Models, Animal ,030104 developmental biology ,Astrocytes ,biology.protein ,Diseases of the nervous system ,lcsh:Q ,Neuron ,Neuroscience ,030217 neurology & neurosurgery - Abstract
Cell-to-cell communications are critical determinants of pathophysiological phenotypes, but methodologies for their systematic elucidation are lacking. Herein, we propose an approach for the Systematic Elucidation and Assessment of Regulatory Cell-to-cell Interaction Networks (SEARCHIN) to identify ligand-mediated interactions between distinct cellular compartments. To test this approach, we selected a model of amyotrophic lateral sclerosis (ALS), in which astrocytes expressing mutant superoxide dismutase-1 (mutSOD1) kill wild-type motor neurons (MNs) by an unknown mechanism. Our integrative analysis that combines proteomics and regulatory network analysis infers the interaction between astrocyte-released amyloid precursor protein (APP) and death receptor-6 (DR6) on MNs as the top predicted ligand-receptor pair. The inferred deleterious role of APP and DR6 is confirmed in vitro in models of ALS. Moreover, the DR6 knockdown in MNs of transgenic mutSOD1 mice attenuates the ALS-like phenotype. Our results support the usefulness of integrative, systems biology approach to gain insights into complex neurobiological disease processes as in ALS and posit that the proposed methodology is not restricted to this biological context and could be used in a variety of other non-cell-autonomous communication mechanisms., Neuron-astrocyte communication plays a key role in pathophysiology, however systematic approaches to unveil it are limited. Here, the authors propose SEARCHIN, a multi-modal integrated workflow, as a tool to identify cross-compartment ligand-receptor interactions, applied to ALS models.
- Published
- 2020
16. Animal Models of CMT2A: State-of-art and Therapeutic Implications
- Author
-
Giacomo P. Comi, Monica Nizzardo, Roberta De Gioia, Stefania Corti, Gaia Citterio, Elena Abati, Federica Rizzo, and Nereo Bresolin
- Subjects
0301 basic medicine ,Transgene ,Neuroscience (miscellaneous) ,MFN2 ,Disease ,Biology ,Gene mutation ,Article ,Animals, Genetically Modified ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,0302 clinical medicine ,CMT2A ,Charcot-Marie-Tooth Disease ,Strengths and weaknesses ,Animals ,Humans ,Animal model ,Clinical Trials as Topic ,Genetic heterogeneity ,Phenotype ,Disease Models, Animal ,030104 developmental biology ,Neurology ,State of art ,Identification (biology) ,Neuroscience ,030217 neurology & neurosurgery - Abstract
Charcot–Marie–Tooth disease type 2A (CMT2A), arising from mitofusin 2 (MFN2) gene mutations, is the most common inherited axonal neuropathy affecting motor and sensory neurons. The cellular and molecular mechanisms by whichMFN2mutations determine neuronal degeneration are largely unclear. No effective treatment exists for CMT2A, which has a high degree of genetic/phenotypic heterogeneity. The identification of mutations inMFN2has allowed the generation of diverse transgenic animal models, but to date, their ability to recapitulate the CMT2A phenotype is limited, precluding elucidation of its pathogenesis and discovery of therapeutic strategies. This review will critically present recent progress in in vivo CMT2A disease modeling, discoveries, drawbacks and limitations, current challenges, and key reflections to advance the field towards developing effective therapies for these patients.
- Published
- 2020
17. Current understanding of and emerging treatment options for spinal muscular atrophy with respiratory distress type 1 (SMARD1)
- Author
-
Giacomo P. Comi, Stefania Corti, Martina G. L. Perego, Monica Nizzardo, Alessandra Govoni, Michela Taiana, Nereo Bresolin, and Noemi Galli
- Subjects
Genetic enhancement ,Cell- and Tissue-Based Therapy ,Disease ,SMN1 ,Bioinformatics ,Muscular Atrophy, Spinal ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,Quality of life (healthcare) ,Neural Stem Cells ,medicine ,Animals ,Humans ,Molecular Biology ,Pharmacology ,Respiratory Distress Syndrome, Newborn ,0303 health sciences ,Respiratory distress ,business.industry ,030302 biochemistry & molecular biology ,Genetic Therapy ,Cell Biology ,Spinal muscular atrophy ,Motor neuron ,medicine.disease ,SMA ,Survival of Motor Neuron 1 Protein ,DNA-Binding Proteins ,medicine.anatomical_structure ,Molecular Medicine ,business ,Ribosomes ,Transcription Factors - Abstract
Spinal muscular atrophy (SMA) with respiratory distress type 1 (SMARD1) is an autosomal recessive motor neuron disease that is characterized by distal and proximal muscle weakness and diaphragmatic palsy that leads to respiratory distress. Without intervention, infants with the severe form of the disease die before 2 years of age. SMARD1 is caused by mutations in the IGHMBP2 gene that determine a deficiency in the encoded IGHMBP2 protein, which plays a critical role in motor neuron survival because of its functions in mRNA processing and maturation. Although it is rare, SMARD1 is the second most common motor neuron disease of infancy, and currently, treatment is primarily supportive. No effective therapy is available for this devastating disease, although multidisciplinary care has been an essential element of the improved quality of life and life span extension in these patients in recent years. The objectives of this review are to discuss the current understanding of SMARD1 through a summary of the presently known information regarding its clinical presentation and pathogenesis and to discuss emerging therapeutic approaches. Advances in clinical care management have significantly extended the lives of individuals affected by SMARD1 and research into the molecular mechanisms that lead to the disease has identified potential strategies for intervention that target the underlying causes of SMARD1. Gene therapy via gene replacement or gene correction provides the potential for transformative therapies to halt or possibly prevent neurodegenerative disease in SMARD1 patients. The recent approval of the first gene therapy approach for SMA associated with mutations in the SMN1 gene may be a turning point for the application of this strategy for SMARD1 and other genetic neurological diseases.
- Published
- 2020
18. Spinal muscular atrophy with respiratory distress type 1: Clinical phenotypes, molecular pathogenesis and therapeutic insights
- Author
-
Nereo Bresolin, Alessandra Govoni, Monica Nizzardo, Giacomo P. Comi, Matteo Saladini, Michela Taiana, and Stefania Corti
- Subjects
0301 basic medicine ,IGHMBP2 ,Genetic enhancement ,Reviews ,Diaphragmatic breathing ,Review ,Disease ,Bioinformatics ,Muscular Atrophy, Spinal ,03 medical and health sciences ,0302 clinical medicine ,medicine ,Animals ,Humans ,Molecular Targeted Therapy ,Respiratory Distress Syndrome, Newborn ,Palsy ,Respiratory distress ,business.industry ,Cell Biology ,Spinal muscular atrophy ,Prognosis ,medicine.disease ,gene therapy ,Phenotype ,DNA-Binding Proteins ,030104 developmental biology ,030220 oncology & carcinogenesis ,Spinal muscular atrophy with respiratory distress type 1 ,Mutation ,motor neuron disease ,Breathing ,Molecular Medicine ,business ,Transcription Factors - Abstract
Spinal muscular atrophy with respiratory distress type 1 (SMARD1) is a rare autosomal recessive neuromuscular disorder caused by mutations in the IGHMBP2 gene, which encodes immunoglobulin μ‐binding protein 2, leading to progressive spinal motor neuron degeneration. We review the data available in the literature about SMARD1. The vast majority of patients show an onset of typical symptoms in the first year of life. The main clinical features are distal muscular atrophy and diaphragmatic palsy, for which permanent supportive ventilation is required. No effective treatment is available yet, but novel therapeutic approaches, such as gene therapy, have shown encouraging results in preclinical settings and thus represent possible methods for treating SMARD1. Significant advancements in the understanding of both the SMARD1 clinical spectrum and its molecular mechanisms have allowed the rapid translation of preclinical therapeutic strategies to human patients to improve the poor prognosis of this devastating disease.
- Published
- 2019
19. Dysregulation of Muscle-Specific MicroRNAs as Common Pathogenic Feature Associated with Muscle Atrophy in ALS, SMA and SBMA: Evidence from Animal Models and Human Patients
- Author
-
Angelo Poletti, Viviana Pensato, Monica Nizzardo, Paola Cavalcante, Cinzia Gellera, Mariarita Galbiati, Silvia Bonanno, Michela Taiana, Davide Pareyson, Giuseppe Lauria, Lorenzo Maggi, Franco Salerno, Stefania Corti, Silvia Fenu, Francesca Andreetta, Anna Venerando, Claudia Malacarne, Renato Mantegazza, Eleonora Dalla Bella, Riccardo Masson, Stefania Marcuzzo, Pia Bernasconi, Cinza Cagnoli, Eleonora Giagnorio, Malacarne, C, Galbiati, M, Giagnorio, E, Cavalcante, P, Salerno, F, Andreetta, F, Cagnoli, C, Taiana, M, Nizzardo, M, Corti, S, Pensato, V, Venerando, A, Gellera, C, Fenu, S, Pareyson, D, Masson, R, Maggi, L, Bella, E, Lauria, G, Mantegazza, R, Bernasconi, P, Poletti, A, Bonanno, S, and Marcuzzo, S
- Subjects
muscle-specific microRNAs ,Pathology ,medicine.medical_specialty ,amyotrophic lateral sclerosis ,motor neuron diseases ,QH301-705.5 ,Muscle-specific microRNA ,Mutation, Missense ,Mice, Transgenic ,Bulbo-Spinal Atrophy, X-Linked ,Catalysis ,Article ,Mouse model ,Inorganic Chemistry ,Pathogenesis ,Mice ,Superoxide Dismutase-1 ,Medicine ,Animals ,Humans ,spinal bulbar muscular atrophy ,mouse models ,Motor neuron disease ,Physical and Theoretical Chemistry ,Amyotrophic lateral sclerosis ,Biology (General) ,Molecular Biology ,QD1-999 ,Spectroscopy ,Amyotrophic lateral sclerosi ,spinal muscular atrophy ,business.industry ,Superoxide Dismutase ,Organic Chemistry ,Skeletal muscle ,General Medicine ,Spinal muscular atrophy ,Motor neuron ,medicine.disease ,SMA ,Muscle atrophy ,Computer Science Applications ,Spinal and bulbar muscular atrophy ,Chemistry ,MicroRNAs ,medicine.anatomical_structure ,Amino Acid Substitution ,medicine.symptom ,business - Abstract
Motor neuron diseases (MNDs) are neurodegenerative disorders characterized by upper and/or lower MN loss. MNDs include amyotrophic lateral sclerosis (ALS), spinal muscular atrophy (SMA), and spinal and bulbar muscular atrophy (SBMA). Despite variability in onset, progression, and genetics, they share a common skeletal muscle involvement, suggesting that it could be a primary site for MND pathogenesis. Due to the key role of muscle-specific microRNAs (myomiRs) in skeletal muscle development, by real-time PCR we investigated the expression of miR-206, miR-133a, miR-133b, and miR-1, and their target genes, in G93A-SOD1 ALS, Δ7SMA, and KI-SBMA mouse muscle during disease progression. Further, we analyzed their expression in serum of SOD1-mutated ALS, SMA, and SBMA patients, to demonstrate myomiR role as noninvasive biomarkers. Our data showed a dysregulation of myomiRs and their targets, in ALS, SMA, and SBMA mice, revealing a common pathogenic feature associated with muscle impairment. A similar myomiR signature was observed in patients’ sera. In particular, an up-regulation of miR-206 was identified in both mouse muscle and serum of human patients. Our overall findings highlight the role of myomiRs as promising biomarkers in ALS, SMA, and SBMA. Further investigations are needed to explore the potential of myomiRs as therapeutic targets for MND treatment.
- Published
- 2021
20. Cell-penetrating peptide-conjugated Morpholino rescues SMA in a symptomatic preclinical model
- Author
-
Mafalda Rizzuti, Margherita Bersani, Manuela Garbellini, Domenica Saccomanno, Giacomo P. Comi, Nereo Bresolin, Elisa Pagliari, Hong M. Moulton, Stefania Corti, and Monica Nizzardo
- Subjects
Pharmacology ,Morpholino ,Spinal muscular atrophy ,Neuropathology ,Cell-Penetrating Peptides ,Motor neuron ,Oligonucleotides, Antisense ,medicine.disease ,SMA ,Phenotype ,Morpholinos ,Muscular Atrophy, Spinal ,Disease Models, Animal ,medicine.anatomical_structure ,Drug Discovery ,Genetics ,Systemic administration ,medicine ,Cell-penetrating peptide ,Cancer research ,Molecular Medicine ,Animals ,Humans ,Molecular Biology - Abstract
Spinal muscular atrophy (SMA) is a motor neuron disease and the leading genetic cause of infant mortality. Recently approved SMA therapies have transformed a deadly disease into a survivable one, but these compounds show a wide spectrum of clinical response and effective rescue only in the early stages of the disease. Therefore, safe, symptomatic-suitable, non-invasive treatments with high clinical impact across different phenotypes are urgently needed. We conjugated antisense oligonucleotides with Morpholino (MO) chemistry, that increase SMN protein levels, to cell-penetrating peptides (CPPs) for better cellular distribution. Systemically administered MOs linked to r6 and (RXRRBR)2XB peptides crossed the blood-brain barrier and increased SMN protein levels remarkably, causing striking improvement of survival, neuromuscular function, and neuropathology, even in symptomatic SMA animals. Our study demonstrates that MO-CPP conjugates can significantly expand the therapeutic window through minimally invasive systemic administration, opening the path for clinical applications of this strategy.
- Published
- 2020
21. Neural Stem Cell Transplantation for Neurodegenerative Diseases
- Author
-
Gaia Citterio, Roberta De Gioia, Giacomo P. Comi, Monica Nizzardo, Elena Abati, Federica Rizzo, Nereo Bresolin, Stefania Corti, and Fabio Biella
- Subjects
Review ,Biology ,Neuroprotection ,Catalysis ,neural subpopulation ,Immunomodulation ,Inorganic Chemistry ,Cell therapy ,lcsh:Chemistry ,neurodegenerative disease ,Neural Stem Cells ,Neurotrophic factors ,medicine ,Animals ,Humans ,Nerve Growth Factors ,Physical and Theoretical Chemistry ,Molecular Biology ,lcsh:QH301-705.5 ,Spectroscopy ,Neuroinflammation ,Organic Chemistry ,Neurodegeneration ,neuronal stem cells ,Neurodegenerative Diseases ,General Medicine ,medicine.disease ,Neural stem cell ,Computer Science Applications ,Transplantation ,nervous system ,lcsh:Biology (General) ,lcsh:QD1-999 ,Stem cell ,cell therapy ,Neuroscience ,Stem Cell Transplantation - Abstract
Neurodegenerative diseases are disabling and fatal neurological disorders that currently lack effective treatment. Neural stem cell (NSC) transplantation has been studied as a potential therapeutic approach and appears to exert a beneficial effect against neurodegeneration via different mechanisms, such as the production of neurotrophic factors, decreased neuroinflammation, enhanced neuronal plasticity and cell replacement. Thus, NSC transplantation may represent an effective therapeutic strategy. To exploit NSCs’ potential, some of their essential biological characteristics must be thoroughly investigated, including the specific markers for NSC subpopulations, to allow profiling and selection. Another key feature is their secretome, which is responsible for the regulation of intercellular communication, neuroprotection, and immunomodulation. In addition, NSCs must properly migrate into the central nervous system (CNS) and integrate into host neuronal circuits, enhancing neuroplasticity. Understanding and modulating these aspects can allow us to further exploit the therapeutic potential of NSCs. Recent progress in gene editing and cellular engineering techniques has opened up the possibility of modifying NSCs to express select candidate molecules to further enhance their therapeutic effects. This review summarizes current knowledge regarding these aspects, promoting the development of stem cell therapies that could be applied safely and effectively in clinical settings.
- Published
- 2020
22. TDP-43 promotes the formation of neuromuscular synapses through the regulation of Disc-large expression in Drosophila skeletal muscles
- Author
-
Giulia Romano, Fabian Feiguin, Monica Nizzardo, Marta Marzullo, Nina Strah, Laura Ciapponi, Aram Megighian, Clelia Introna, and Raffaella Klima
- Subjects
TDP-43 ,Physiology ,Plant Science ,0302 clinical medicine ,Structural Biology ,RNA interference ,Drosophila Proteins ,Skeletal muscles ,Amyotrophic lateral sclerosis ,lcsh:QH301-705.5 ,Motor Neurons ,0303 health sciences ,Pathophysiology ,Cell biology ,DNA-Binding Proteins ,Drosophila melanogaster ,Dlg ,medicine.anatomical_structure ,General Agricultural and Biological Sciences ,Intracellular ,Research Article ,Biotechnology ,Biology ,General Biochemistry, Genetics and Molecular Biology ,03 medical and health sciences ,Cell Line, Tumor ,mental disorders ,medicine ,Animals ,Humans ,Muscle, Skeletal ,Ecology, Evolution, Behavior and Systematics ,030304 developmental biology ,fungi ,Amyotrophic Lateral Sclerosis ,nutritional and metabolic diseases ,Skeletal muscle ,Cell Biology ,biology.organism_classification ,medicine.disease ,ALS ,neuromuscular junctions ,skeletal muscles ,nervous system diseases ,lcsh:Biology (General) ,Synapses ,Neuromuscular junctions ,030217 neurology & neurosurgery ,Function (biology) ,Homeostasis ,Developmental Biology - Abstract
Background The ribonuclear protein TDP-43 has been implicated in the pathophysiology of amyotrophic lateral sclerosis (ALS), with genetic mutations being linked to the neurological symptoms of the disease. Though alterations in the intracellular distribution of TDP-43 have been observed in skeletal muscles of patients suffering from ALS, it is not clear whether such modifications play an active role in the disease or merely represent an expression of muscle homeostatic mechanisms. Also, the molecular and metabolic pathways regulated by TDP-43 in the skeletal muscle remain largely unknown. Here, we analyze the function of TBPH, the Drosophila melanogaster ortholog of TDP-43, in skeletal muscles. Results We modulated the activity of TDP-43 in Drosophila muscles by means of RNA interference and observed that it is required to promote the formation and growth of neuromuscular synapses. TDP-43 regulated the expression levels of Disc-large (Dlg), and restoring Dlg expression either in skeletal muscles or in motoneurons was sufficient to suppress the locomotive and synaptic defects of TDP-43-null flies. These results were validated by the observation of a decrease in Dlg levels in human neuroblastoma cells and iPSC-differentiated motoneurons derived from ALS patients, suggesting similar mechanisms may potentially be involved in the pathophysiology of the disease. Conclusions Our results help to unveil the physiological role of TDP-43 in skeletal muscles as well as the mechanisms responsible for the autonomous and non-autonomous behavior of this protein concerning the organization of neuromuscular synapses.
- Published
- 2020
23. miR-129-5p: A key factor and therapeutic target in amyotrophic lateral sclerosis
- Author
-
Christian Lunetta, Oliver Mühlemann, Carlo Ferrarese, Silvia M.L. Barabino, Antonella Ronchi, Lucio Tremolizzo, Raffaele A. Calogero, Alessia Loffreda, Caterina Bendotti, Monica Nizzardo, Marc-David Ruepp, Stefano Volinia, Stefania Corti, Marco Galasso, A Arosio, Mafalda Rizzuti, Loffreda, A, Nizzardo, M, Arosio, A, Ruepp, M, Calogero, R, Volinia, S, Galasso, M, Bendotti, C, Ferrarese, C, Lunetta, C, Rizzuti, M, Ronchi, A, Mühlemann, O, Tremolizzo, L, Corti, S, and Barabino, S
- Subjects
0301 basic medicine ,Small RNA ,Neurite ,Therapeutic target ,SOD1 ,SOD-1 ,Oligonucleotides ,Down-Regulation ,ELAV-Like Protein 4 ,Biology ,03 medical and health sciences ,chemistry.chemical_compound ,Mice ,0302 clinical medicine ,Superoxide Dismutase-1 ,Downregulation and upregulation ,540 Chemistry ,microRNA ,medicine ,Gene silencing ,Animals ,Humans ,Antagomir ,Amyotrophic lateral sclerosis ,Antisense ,miRNA ,MED/26 - NEUROLOGIA ,Animal ,General Neuroscience ,Amyotrophic Lateral Sclerosis ,Oligonucleotides, Antisense ,medicine.disease ,BIO/11 - BIOLOGIA MOLECOLARE ,Up-Regulation ,Disease Models, Animal ,MicroRNAs ,030104 developmental biology ,chemistry ,Disease Models ,Cancer research ,570 Life sciences ,biology ,ALS ,Neuroscience ,030217 neurology & neurosurgery - Abstract
Amyotrophic lateral sclerosis (ALS) is a relentless and fatal neurological disease characterized by the selective degeneration of motor neurons. No effective therapy is available for this disease. Several lines of evidence indicate that alteration of RNA metabolism, including microRNA (miRNA) processing, is a relevant pathogenetic factor and a possible therapeutic target for ALS. Here, we showed that the abundance of components in the miRNA processing machinery is altered in a SOD1-linked cellular model, suggesting consequent dysregulation of miRNA biogenesis. Indeed, high-throughput sequencing of the small RNA fraction showed that among the altered miRNAs, miR-129-5p was increased in different models of SOD1-linked ALS and in peripheral blood cells of sporadic ALS patients. We demonstrated that miR-129-5p upregulation causes the downregulation of one of its targets: the RNA-binding protein ELAVL4/HuD. ELAVL4/HuD is predominantly expressed in neurons, where it controls several key neuronal mRNAs. Overexpression of pre-miR-129-1 inhibited neurite outgrowth and differentiation via HuD silencing in vitro, while its inhibition with an antagomir rescued the phenotype. Remarkably, we showed that administration of an antisense oligonucleotide (ASO) inhibitor of miR-129-5p to an ALS animal model, SOD1 (G93A) mice, result in a significant increase in survival and improved the neuromuscular phenotype in treated mice. These results identify miR-129-5p as a therapeutic target that is amenable to ASO modulation for the treatment of ALS patients.
- Published
- 2020
24. Natural history study of spinal muscular atrophy with respiratory distress type 1 (SMARD1) in a cohort of European patients
- Author
-
Monica Nizzardo, Enrico Bertini, Stefania Corti, Adele D'Amico, Joaquín Alejandro Fernández-Ramos, Emilio Albamonte, Sonia Messina, Liliana Porfiri, Magdalena Piontek, Alessandra Govoni, Michela Taiana, Francesco Danilo Tiziano, Giacomo P. Comi, Gianluca Vita, Elisabetta Cesaroni, Francesco Mari, Andi Nuredini, Iwona Ostrowska, Maria Sframeli, and Valeria A. Sansone
- Subjects
medicine.medical_specialty ,Neurology ,Respiratory distress ,business.industry ,Internal medicine ,Cohort ,medicine ,Neurology (clinical) ,Spinal muscular atrophy ,medicine.disease ,business ,Natural history study - Published
- 2021
25. Dysregulation of myomiRs as common pathogenic feature associated with muscle atrophy in ALS, SMA and SBMA: Evidence from animal models and human patients
- Author
-
Monica Nizzardo, Viviana Pensato, Silvia Bonanno, Michela Taiana, Giuseppe Lauria, Riccardo Masson, Eleonora Giagnorio, Lorenzo Maggi, Renato Mantegazza, Cinzia Gellera, Silvia Fenu, Francesca Andreetta, Paola Cavalcante, Franco Salerno, Davide Pareyson, Stefania Corti, Claudia Malacarne, Eleonora Dalla Bella, Mariarita Galbiati, Cinzia Cagnoli, Stefania Marcuzzo, Angelo Poletti, and Anna Venerando
- Subjects
Neurology ,Feature (computer vision) ,business.industry ,medicine ,Neurology (clinical) ,medicine.symptom ,SMA ,business ,Neuroscience ,Muscle atrophy - Published
- 2021
26. Human spinal cord-like organoids to model C9ORF72 ALS and test new therapies in vitro
- Author
-
Monica Nizzardo, Irene Faravelli, Stefano Ghezzi, Stefania Corti, Gianluca Costamagna, Michela Taiana, Noemi Galli, Mafalda Rizzuti, Benedetta Frizzi, Fabio Biella, and Giacomo P. Comi
- Subjects
Pathology ,medicine.medical_specialty ,medicine.anatomical_structure ,Neurology ,business.industry ,C9orf72 ,medicine ,Organoid ,Neurology (clinical) ,Spinal cord ,business ,In vitro - Published
- 2021
27. Synaptotagmin 13 is neuroprotective across motor neuron diseases
- Author
-
Giacomo P. Comi, Monica Nizzardo, J. Aguila Benitez, Ilary Allodi, Stefania Corti, Valentina Melzi, Nereo Bresolin, Jik Nijssen, Eva Hedlund, Federica Rizzo, and Michela Taiana
- Subjects
Male ,Mice, Transgenic ,Biology ,Neuroprotection ,Synaptotagmin 1 ,Pathology and Forensic Medicine ,Cellular and Molecular Neuroscience ,Mice ,Synaptotagmins ,medicine ,Animals ,Humans ,Amyotrophic lateral sclerosis ,Axon ,Motor Neuron Disease ,Motor Neurons ,Original Paper ,Superoxide Dismutase ,Amyotrophic Lateral Sclerosis ,Spinal muscular atrophy ,Motor neuron ,medicine.disease ,SMA ,Disease Models, Animal ,medicine.anatomical_structure ,nervous system ,Female ,Neurology (clinical) ,Brainstem ,Neuroscience - Abstract
In amyotrophic lateral sclerosis (ALS) and spinal muscular atrophy (SMA), spinal and lower brainstem motor neurons degenerate, but some motor neuron subtypes are spared, including oculomotor neurons (OMNs). The mechanisms responsible for this selective degeneration are largely unknown, but the molecular signatures of resistant and vulnerable motor neurons are distinct and offer clues to neuronal resilience and susceptibility. Here, we demonstrate that healthy OMNs preferentially express Synaptotagmin 13 (SYT13) compared to spinal motor neurons. In end-stage ALS patients, SYT13 is enriched in both OMNs and the remaining relatively resilient spinal motor neurons compared to controls. Overexpression of SYT13 in ALS and SMA patient motor neurons in vitro improves their survival and increases axon lengths. Gene therapy with Syt13 prolongs the lifespan of ALS mice by 14% and SMA mice by 50% by preserving motor neurons and delaying muscle denervation. SYT13 decreases endoplasmic reticulum stress and apoptosis of motor neurons, both in vitro and in vivo. Thus, SYT13 is a resilience factor that can protect motor neurons and a candidate therapeutic target across motor neuron diseases. Electronic supplementary material The online version of this article (10.1007/s00401-020-02133-x) contains supplementary material, which is available to authorized users.
- Published
- 2019
28. TDP-43 regulates the expression levels of Disc-large in skeletal muscles to promote the assemble of the neuromuscular synapses in Drosophila
- Author
-
Aram Megighian, Fabian Feiguin, Raffaella Klima, Strah N, Giulia Romano, Introna C, and Monica Nizzardo
- Subjects
biology ,Human cell ,biology.organism_classification ,Drosophila ,Homeostasis ,Intracellular ,Cell biology - Abstract
BackgroundAlterations in the intracellular distribution of TDP-43 were observed in the skeletal muscles of patients suffering from ALS. However, it is not clear whether these modifications play an active role in the disease or represent a physiological adaptation to muscles homeostasis.ResultTo answer these questions, we modulated the activity of this protein in Drosophila muscles and observed that TDP-43 was required in these tissues to promote the formation and growth of the neuromuscular synapses. Moreover, we identified that TDP-43 regulates the expression levels of Disc-large (Dlg) and demonstrated that the modulation of Dlg activity, in skeletal muscles or motoneurons, was sufficient to recover the TDP-43-null locomotive and synaptic defects in flies. Additionally, we found that similar mechanisms are conserved in human cell lines and present in tissues derived from ALS patients.ConclusionsOur results uncover the physiological role of TDP-43 in skeletal muscles as well as the mechanisms behind the autonomous and non-autonomous behavior of this protein in the organization of the neuromuscular synapses.
- Published
- 2019
29. MicroRNA expression analysis identifies a subset of downregulated miRNAs in ALS motor neuron progenitors
- Author
-
Mafalda Rizzuti, Giuseppe Filosa, Luca Calandriello, Nereo Bresolin, Giacomo P. Comi, Silvia M.L. Barabino, Stefania Corti, Valentina Melzi, Monica Nizzardo, Valentina Bollati, Laura Dioni, Rizzuti, M, Filosa, G, Melzi, V, Calandriello, L, Dioni, L, Bollati, V, Bresolin, N, Comi, G, Barabino, S, Nizzardo, M, and Corti, S
- Subjects
0301 basic medicine ,Induced Pluripotent Stem Cells ,Gene regulatory network ,lcsh:Medicine ,Down-Regulation ,Biology ,Article ,03 medical and health sciences ,Neural Stem Cells ,microRNA ,medicine ,Humans ,Gene Regulatory Networks ,Amyotrophic lateral sclerosis ,Progenitor cell ,Induced pluripotent stem cell ,lcsh:Science ,Gene ,Cells, Cultured ,Motor Neurons ,Multidisciplinary ,lcsh:R ,Amyotrophic Lateral Sclerosis ,Motor neuron ,medicine.disease ,Phenotype ,Cell biology ,MicroRNAs ,030104 developmental biology ,medicine.anatomical_structure ,lcsh:Q ,Synaptic Vesicles ,Tumor Suppressor Protein p53 - Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal neurological disorder that is characterized by a progressive degeneration of motor neurons (MNs). The pathomechanism underlying the disease is largely unknown, even though increasing evidence suggests that RNA metabolism, including microRNAs (miRNAs) may play an important role. In this study, human ALS induced pluripotent stem cells were differentiated into MN progenitors and their miRNA expression profiles were compared to those of healthy control cells. We identified 15 downregulated miRNAs in patients’ cells. Gene ontology and molecular pathway enrichment analysis indicated that the predicted target genes of the differentially expressed miRNAs were involved in neurodegeneration-related pathways. Among the 15 examined miRNAs, miR-34a and miR504 appeared particularly relevant due to their involvement in the p53 pathway, synaptic vesicle regulation and general involvement in neurodegenerative diseases. Taken together our results demonstrate that the neurodegenerative phenotype in ALS can be associated with a dysregulation of miRNAs involved in the control of disease-relevant genetic pathways, suggesting that targeting entire gene networks can be a potential strategy to treat complex diseases such as ALS.
- Published
- 2018
30. Revealing the involvement of miR-376a, miR-432 and miR-451a in infantile ascending hereditary spastic paralysis by microRNA profiling in iPSCs
- Author
-
Sara D’Alessandro, Claudia Barzago, Giovanna Zorzi, Massimo Mantegazza, Paola Cavalcante, Stefania Corti, Michela Taiana, Claudia Malacarne, Monica Nizzardo, Renato Mantegazza, Antonio Gambardella, Silvia Bonanno, Barbara Galbardi, Silvana Franceschetti, Giulia Bechi, Stefania Marcuzzo, Pia Bernasconi, Marcuzzo, S, Bonanno, S, Barzago, C, D’Alessandro, S, Cavalcante, P, Galbardi, B, Malacarne, C, Taiana, M, Nizzardo, M, Corti, S, Bechi, G, Gambardella, A, Franceschetti, S, Mantegazza, M, Zorzi, G, Mantegazza, R, and Bernasconi, P
- Subjects
business.industry ,alsin, induced pluripotent stem cells, infantile-onset ascending, hereditary spastic paralysis, microRNAs, motor neuron disease ,Cancer research ,Medicine ,business ,Induced pluripotent stem cell ,Microrna profiling ,Infantile ascending hereditary spastic paralysis - Abstract
Infantile-onset ascending hereditary spastic paralysis (IAHSP) is a rare, early onset, autosomal recessive motor neuron disease characterized by progressive weakness and spasticity. Several mutations in the alsin 2 gene (ALS2) have been described in IAHSP patients; however, a relevant subset of patients is ALS2 mutation-negative, and pathogenic events causing the disease are unknown. The present study aimed at better understanding the molecular mechanisms underlying motor neuron loss in IAHSP patients by identifying microRNAs (miRNAs) potentially implicated in neuronal differentiation. Using the human induced pluripotent stem cell (iPSC) technology, we developed a patient-specific in vitro cellular model and performed miRNome profiling in fibroblasts, iPSCs and iPSCs-derived neurons obtained from an ALS2 mutation-negative IAHSP patient and a healthy control. The selected differentially expressed miRNAs were also analyzed in fibroblasts, iPSCs and iPSCs-derived neurons from two patients affected by other motor neuron diseases, two patients with other neurological disease, and three healthy controls. We found that miR-376a, miR-432 and miR-451a expression was altered in cell cultures obtained from the IAHSP patient compared to the other patients and controls. In addition, the hierarchical clustering analysis revealed that miR-451a was differentially expressed in fibroblasts and iPSCs, whereas miR-376a and miR-432 in neuronal cells. These results, together with the miRNA/mRNA target analysis, were indicative of a significant involvement of miR-451a in stem cell biology processes, and of miR-376a and miR-432 in the establishment of the neuronal phenotype. Our overall findings identified miR-376a, miR-432 and miR-451a as molecules involved in neuronal differentiation, and potentially in IAHSP pathogenesis, which could provide cues for future development of patient-specific miRNA-based therapeutic strategies for IAHSP or other motor neuron diseases
- Published
- 2018
31. Mitochondrial Dysregulation and Impaired Autophagy in iPSC-Derived Dopaminergic Neurons of Multiple System Atrophy
- Author
-
Monica Nizzardo, Maura Samarani, Alessio Di Fonzo, Rosamaria Silipigni, Giacomo P. Comi, Maria Passafaro, Emanuele Frattini, Christian Bergamini, Manuela Garbellini, Giacomo Monzio Compagnoni, Arianna Bellucci, Massimo Aureli, Catarina M. Quinzii, Romana Fato, Francesco Fortunato, Sabrina Salani, Stefania Corti, Dario Ronchi, Elena Abati, Andreina Bordoni, Giulio Kleiner, Giulia Maia Serratto, Nereo Bresolin, Monica Miozzo, Michela Deleidi, Silvia Tabano, Gaia Faustini, Monzio Compagnoni, Giacomo, Kleiner, Giulio, Samarani, Maura, Aureli, Massimo, Faustini, Gaia, Bellucci, Arianna, Ronchi, Dario, Bordoni, Andreina, Garbellini, Manuela, Salani, Sabrina, Fortunato, Francesco, Frattini, Emanuele, Abati, Elena, Bergamini, Christian, Fato, Romana, Tabano, Silvia, Miozzo, Monica, Serratto, Giulia, Passafaro, Maria, Deleidi, Michela, Silipigni, Rosamaria, Nizzardo, Monica, Bresolin, Nereo, Comi, Giacomo P., Corti, Stefania, Quinzii, Catarina M., and Di Fonzo, Alessio
- Subjects
0301 basic medicine ,Male ,induced pluripotent stem cell ,pathology [Dopaminergic Neurons] ,Respiratory chain ,multiple system atrophy ,Mitochondrion ,Biochemistry ,pathology [Mitochondria] ,Pathogenesis ,0302 clinical medicine ,pathology [Multiple System Atrophy] ,Induced pluripotent stem cell ,lcsh:QH301-705.5 ,Aged, 80 and over ,lcsh:R5-920 ,Neurodegeneration ,Dopaminergic ,neurodegeneration ,Middle Aged ,pathology [Induced Pluripotent Stem Cells] ,Mitochondria ,Female ,lcsh:Medicine (General) ,metabolism [Biomarkers] ,autophagy ,MSA ,dopaminergic neurons ,induced pluripotent stem cells ,mitochondria ,Biology ,Article ,03 medical and health sciences ,Young Adult ,Atrophy ,Genetic ,dopaminergic neuron ,Genetics ,medicine ,Autophagy ,Humans ,ddc:610 ,Aged ,Cell Biology ,medicine.disease ,030104 developmental biology ,nervous system ,lcsh:Biology (General) ,Case-Control Studies ,Neuroscience ,030217 neurology & neurosurgery ,Biomarkers ,Developmental Biology - Abstract
Summary Multiple system atrophy (MSA) is a progressive neurodegenerative disease that affects several areas of the CNS, whose pathogenesis is still widely unclear and for which an effective treatment is lacking. We have generated induced pluripotent stem cell-derived dopaminergic neurons from four MSA patients and four healthy controls and from two monozygotic twins discordant for the disease. In this model, we have demonstrated an aberrant autophagic flow and a mitochondrial dysregulation involving respiratory chain activity, mitochondrial content, and CoQ10 biosynthesis. These defective mechanisms may contribute to the onset of the disease, representing potential therapeutic targets., Highlights • An iPSC-based neuronal model of MSA is described • Mitochondria are dysfunctional in MSA neurons • Autophagic machinery is impaired in MSA neurons, Monzio Compagnoni et al. present an iPSC-based neuronal in vitro model of multiple system atrophy. Patients' dopaminergic neurons display a dysregulation of mitochondrial functioning and autophagy, suggesting new hints for the comprehension of the pathogenesis of the disease.
- Published
- 2018
32. Downregulation of glutamic acid decarboxylase in Drosophila TDP-43-null brains provokes paralysis by affecting the organization of the neuromuscular synapses
- Author
-
Federica Grilli, Federica Rizzo, Fabian Feiguin, Rodolfo C. Garcia, Giulia Romano, Monica Nizzardo, Corrado Guarnaccia, Nikola Holodkov, and Raffaella Klima
- Subjects
0301 basic medicine ,Glutamate decarboxylase ,Mutant ,Excitotoxicity ,Neuromuscular Junction ,lcsh:Medicine ,Down-Regulation ,Glutamic Acid ,Biology ,Motor Activity ,medicine.disease_cause ,GAD1 ,Article ,Cell Line ,03 medical and health sciences ,0302 clinical medicine ,Downregulation and upregulation ,medicine ,Animals ,Drosophila Proteins ,Humans ,Paralysis ,Amyotrophic lateral sclerosis ,lcsh:Science ,Receptor ,Motor Neurons ,Multidisciplinary ,Glutamate Decarboxylase ,lcsh:R ,fungi ,Amyotrophic Lateral Sclerosis ,Glutamate receptor ,Brain ,medicine.disease ,Cell biology ,DNA-Binding Proteins ,030104 developmental biology ,Receptors, Glutamate ,Mutation ,Synapses ,lcsh:Q ,Drosophila ,Neuroglia ,030217 neurology & neurosurgery ,Locomotion - Abstract
Amyotrophic lateral sclerosis is a progressive neurodegenerative disease that affects the motor system, comprised of motoneurons and associated glia. Accordingly, neuronal or glial defects in TDP-43 function provoke paralysis due to the degeneration of the neuromuscular synapses in Drosophila. To identify the responsible molecules and mechanisms, we performed a genome wide proteomic analysis to determine differences in protein expression between wild-type and TDP-43-minus fly heads. The data established that mutant insects presented reduced levels of the enzyme glutamic acid decarboxylase (Gad1) and increased concentrations of extracellular glutamate. Genetic rescue of Gad1 activity in neurons or glia was sufficient to recuperate flies locomotion, synaptic organization and glutamate levels. Analogous recovery was obtained by treating TDP-43-null flies with glutamate receptor antagonists demonstrating that Gad1 promotes synapses formation and prevents excitotoxicity. Similar suppression of TDP-43 provoked the downregulation of GAD67, the Gad1 homolog protein in human neuroblastoma cell lines and analogous modifications were observed in iPSC-derived motoneurons from patients carrying mutations in TDP-43, uncovering conserved pathological mechanisms behind the disease.
- Published
- 2018
33. MFN2-related neuropathies: Clinical features, molecular pathogenesis and therapeutic perspectives
- Author
-
Giulia Stuppia, Roberto Del Bo, Giacomo P. Comi, Chiara Simone, Stefania Corti, Giulietta Riboldi, Nereo Bresolin, Monica Nizzardo, and Federica Rizzo
- Subjects
Neurons ,Genetics ,Neurodegeneration ,MFN2 ,Disease ,Biology ,medicine.disease ,GTP Phosphohydrolases ,Mitochondria ,Mitochondrial Proteins ,Pathogenesis ,Mitofusin-2 ,Phenotype ,Neurology ,mitochondrial fusion ,Charcot-Marie-Tooth Disease ,Mutation ,Mitophagy ,medicine ,Animals ,Humans ,Neurology (clinical) ,Neuroscience ,Gene - Abstract
Mitofusin 2 (MFN2) is a GTPase dynamin-like protein of the outer mitochondrial membrane, encoded in the nuclear genome by the MFN2 gene located on the short (p) arm of chromosome 1. MFN2 protein is involved in several intracellular pathways, but is mainly involved in a network that has an essential role in several mitochondrial functions, including fusion, axonal transport, interorganellar communication and mitophagy. Mutations in the gene encoding MFN2 are associated with Charcot-Marie-Tooth disease type 2A (CMT2A), a neurological disorder characterized by a wide clinical phenotype that involves the central and peripheral nervous system. Here, we present the clinical, genetic and neuropathological features of human diseases associated with MFN2 mutations. We also report proposed pathogenic mechanisms through which MFN2 mutations likely contribute to the development of neurodegeneration. MFN2-related disorders may occur more frequently than previously considered, and they may represent a paradigm for the study of the defective mitochondrial dynamics that seem to play a significant role in the molecular and cellular pathogenesis of common neurodegenerative diseases; thus they may also lead to the identification of related therapeutic targets.
- Published
- 2015
34. Spinal muscular atrophy—recent therapeutic advances for an old challenge
- Author
-
Irene Faravelli, Giacomo P. Comi, Stefania Corti, and Monica Nizzardo
- Subjects
Motor Neurons ,Clinical Trials as Topic ,business.industry ,Genetic enhancement ,SMN Complex Proteins ,Survival of motor neuron ,Spinal muscular atrophy ,Disease ,Oligonucleotides, Antisense ,medicine.disease ,SMA ,Diagnosis, Differential ,Muscular Atrophy, Spinal ,Clinical trial ,Transplantation ,Cellular and Molecular Neuroscience ,Animals ,Humans ,Medicine ,Neurology (clinical) ,Stem cell ,business ,Neuroscience - Abstract
In the past decade, improved understanding of spinal muscular atrophy (SMA) aetiopathogenesis has brought us to a historical turning point: we are at the verge of development of disease-modifying treatments for this hitherto incurable disease. The increasingly precise delineation of molecular targets within the survival of motor neuron (SMN) gene locus has led to the development of promising therapeutic strategies. These novel avenues in treatment for SMA include gene therapy, molecular therapy with antisense oligonucleotides, and small molecules that aim to increase expression of SMN protein. Stem cell studies of SMA have provided an in vitro model for SMA, and stem cell transplantation could be used as a complementary strategy with a potential to treat the symptomatic phases of the disease. Here, we provide an overview of established data and novel insights into SMA pathogenesis, including discussion of the crucial function of the SMN protein. Preclinical evidence and recent advances from ongoing clinical trials are thoroughly reviewed. The final remarks are dedicated to future clinical perspectives in this rapidly evolving field, with a broad discussion on the comparison between the outlined therapeutic approaches and the remaining open questions.
- Published
- 2015
35. Experimental Advances Towards Neural Regeneration from Induced Stem Cells to Direct In Vivo Reprogramming
- Author
-
Monica Nizzardo, Agnese Ramirez, Stefania Corti, Margherita Ruggieri, Sara Dametti, and Irene Faravelli
- Subjects
0301 basic medicine ,Induced stem cells ,Regeneration (biology) ,Induced Pluripotent Stem Cells ,Neuroscience (miscellaneous) ,Clinical uses of mesenchymal stem cells ,Biology ,Cellular Reprogramming ,Neural stem cell ,Nerve Regeneration ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,030104 developmental biology ,Neural Stem Cells ,Neurology ,Animals ,Humans ,Stem cell ,Progenitor cell ,Induced pluripotent stem cell ,Neuroscience ,Reprogramming - Abstract
Neuronal loss is a common substrate of many neurological diseases that still lack effective treatments and highly burden lives of affected individuals. The discovery of self-renewing stem cells within the central nervous system (CNS) has opened the doors to the possibility of using the plasticity of CNS as a potential strategy for the development of regenerative therapies after injuries. The role of neural progenitor cells appears to be crucial, but insufficient in reparative processes after damage. In addition, the mechanisms that regulate these events are still largely unknown. Stem cell-based therapeutic approaches have primarily focused on the use of either induced pluripotent stem cells or induced neural stem cells as sources for cell transplantation. More recently, in vivo direct reprogramming of endogenous CNS cells into multipotent neural stem/progenitor cells has been proposed as an alternative strategy that could overcome the limits connected with both the invasiveness of exogenous cell transplantation and the technical issues of in vitro reprogramming (i.e., the time requested and the limited available amount of directly induced neuronal cells). In this review, we aim to highlight the recent studies on in vivo direct reprogramming, focusing on astrocytes conversion to neurons or to neural stem/precursors cells, in the perspective of future therapeutic purposes for neurological disorders.
- Published
- 2015
36. Motor neurons with differential vulnerability to degeneration show distinct protein signatures in health and ALS
- Author
-
Chiara Simone, Eva Hedlund, Monica Nizzardo, Ilary Allodi, Stefania Corti, Susanne Nichterwitz, and Laura H. Comley
- Subjects
Male ,Motor neuron ,Neuroscience(all) ,Dynein ,Cell Count ,Mice, Transgenic ,Selective vulnerability ,Biology ,Oculomotor nucleus ,Anterior Horn Cells ,selective vulnerability ,medicine ,Animals ,Humans ,Premovement neuronal activity ,Amyotrophic lateral sclerosis ,Neurodegeneration ,motor neuron ,Aged ,Aged, 80 and over ,Motor Neurons ,General Neuroscience ,Amyotrophic Lateral Sclerosis ,GUCY1A3 ,neurodegeneration ,oculomotor ,Peripherin ,Middle Aged ,medicine.disease ,Mice, Inbred C57BL ,Disease Models, Animal ,medicine.anatomical_structure ,nervous system ,Nerve Degeneration ,Female ,Neuroscience ,Brain Stem ,Oculomotor - Abstract
The lethal disease amyotrophic lateral sclerosis (ALS) is characterized by the loss of somatic motor neurons. However, not all motor neurons are equally vulnerable to disease; certain groups are spared, including those in the oculomotor nucleus controlling eye movement. The reasons for this differential vulnerability remain unknown. Here we have identified a protein signature for resistant oculomotor motor neurons and vulnerable hypoglossal and spinal motor neurons in mouse and man and in health and ALS with the aim of understanding motor neuron resistance. Several proteins with implications for motor neuron resistance, including GABAA receptor α1, guanylate cyclase soluble subunit alpha-3 and parvalbumin were persistently expressed in oculomotor neurons in man and mouse. Vulnerable motor neurons displayed higher protein levels of dynein, peripherin and GABAA receptor α2, which play roles in retrograde transport and excitability, respectively. These were dynamically regulated during disease and thus could place motor neurons at an increased risk. From our analysis is it evident that oculomotor motor neurons have a distinct protein signature compared to vulnerable motor neurons in brain stem and spinal cord, which could in part explain their resistance to degeneration in ALS. Our comparison of human and mouse shows the relative conservation of signals across species and infers that transgenic SOD1G93A mice could be used to predict mechanisms of neuronal vulnerability in man.
- Published
- 2015
- Full Text
- View/download PDF
37. Therapeutic applications of the cell-penetrating HIV-1 Tat peptide
- Author
-
Monica Nizzardo, Mafalda Rizzuti, Stefania Corti, Chiara Zanetta, and Agnese Ramirez
- Subjects
Biodistribution ,media_common.quotation_subject ,Cell ,Peptide ,Cell-Penetrating Peptides ,Computational biology ,Pharmacology ,Hiv 1 tat ,Antibodies ,Drug Delivery Systems ,Drug conjugation ,Nucleic Acids ,Drug Discovery ,Animals ,Humans ,Medicine ,Internalization ,media_common ,chemistry.chemical_classification ,business.industry ,Oligonucleotides, Antisense ,Clinical therapy ,medicine.anatomical_structure ,Pharmaceutical Preparations ,chemistry ,Biological target ,HIV-1 ,Nanoparticles ,tat Gene Products, Human Immunodeficiency Virus ,business - Abstract
Over the past decades, many new therapeutic approaches have been developed for several conditions, including neurodegenerative diseases. However, efficient biodistribution and delivery at biological target sites are hampered by the presence of cell and tissue barriers, and a clinical therapy is prevented by the requirement of invasive administration routes. Candidate drug conjugation to cell-penetrating peptides, which are able to cross cellular membranes and reach biological targets even when administered systemically, represents a promising tool to overcome this issue. Here, we review the biology, classification and mechanisms of internalization of cell-penetrating peptides. We focus our attention on the cell-penetrating peptide: HIV-derived Tat peptide, and discuss its efficient but controversial use in basic, preclinical and clinical research from its discovery to the present day.
- Published
- 2015
38. Genome-wide RNA-seq of iPSC-derived motor neurons indicates selective cytoskeletal perturbation in Brown–Vialetto disease that is partially rescued by riboflavin
- Author
-
Agnese Ramirez, Giacomo P. Comi, Alessia Niceforo, Claudia Compagnucci, Andreina Bordoni, Francesco Fortunato, Stefania Corti, Sabrina Salani, Nereo Bresolin, Enrico Bertini, Valentina Melzi, Federica Rizzo, and Monica Nizzardo
- Subjects
0301 basic medicine ,Neurofilament ,Hearing Loss, Sensorineural ,Riboflavin ,Bulbar Palsy, Progressive ,Induced Pluripotent Stem Cells ,Neuronal Outgrowth ,Biology ,Article ,Transcriptome ,03 medical and health sciences ,medicine ,Autophagy ,Humans ,Axon ,Induced pluripotent stem cell ,Cells, Cultured ,Cytoskeleton ,Regulation of gene expression ,Motor Neurons ,Multidisciplinary ,Gene Expression Profiling ,Neurodegeneration ,RNA ,High-Throughput Nucleotide Sequencing ,Cell Differentiation ,Motor neuron ,medicine.disease ,Cell biology ,Mitochondria ,030104 developmental biology ,medicine.anatomical_structure ,Neuroprotective Agents ,Biochemistry ,Gene Expression Regulation ,Dietary Supplements ,Energy Metabolism ,Genome-Wide Association Study - Abstract
Riboflavin is essential in numerous cellular oxidation/reduction reactions but is not synthesized by mammalian cells. Riboflavin absorption occurs through the human riboflavin transporters RFVT1 and RFVT3 in the intestine and RFVT2 in the brain. Mutations in these genes are causative for the Brown–Vialetto–Van Laere (BVVL), childhood-onset syndrome characterized by a variety of cranial nerve palsies as well as by spinal cord motor neuron (MN) degeneration. Why mutations in RFVTs result in a neural cell–selective disorder is unclear. As a novel tool to gain insights into the pathomechanisms underlying the disease, we generated MNs from induced pluripotent stem cells (iPSCs) derived from BVVL patients as an in vitro disease model. BVVL-MNs explained a reduction in axon elongation, partially improved by riboflavin supplementation. RNA sequencing profiles and protein studies of the cytoskeletal structures showed a perturbation in the neurofilament composition in BVVL-MNs. Furthermore, exploring the autophagy–lysosome pathway, we observed a reduced autophagic/mitophagic flux in patient MNs. These features represent emerging pathogenetic mechanisms in BVVL-associated neurodegeneration, partially rescued by riboflavin supplementation. Our data showed that this therapeutic strategy could have some limits in rescuing all of the disease features, suggesting the need to develop complementary novel therapeutic strategies.
- Published
- 2017
39. Glycogen storage disease type III: A novel Agl knockout mouse model
- Author
-
Raffaella Violano, Andreina Bordoni, Stefano Gatti, Stefania Corti, Gianna Ulzi, Serena Pagliarani, Michela Ripolone, Sabrina Lucchiari, Giacomo P. Comi, Maurizio Moggio, Nereo Bresolin, and Monica Nizzardo
- Subjects
medicine.medical_specialty ,Cirrhosis ,Glycogen ,Glycogen storage disease type III ,Glycogenosis ,Cardiomyopathy ,Metabolic disease ,Exercise intolerance ,Hypoglycemia ,Biology ,medicine.disease ,Mouse model ,Glycogen debranching enzyme ,chemistry.chemical_compound ,Endocrinology ,chemistry ,Internal medicine ,Knockout mouse ,medicine ,Molecular Medicine ,medicine.symptom ,Molecular Biology - Abstract
Glycogen storage disease type III is an autosomal recessive disease characterized by a deficiency in the glycogen debranching enzyme, encoded by AGL. Essential features of this disease are hepatomegaly, hypoglycemia, hyperlipidemia, and growth retardation. Progressive skeletal myopathy, neuropathy, and/or cardiomyopathy become prominent in adults. Currently, there is no available cure. We generated an Agl knockout mouse model by deletion of the carboxy terminus of the protein, including the carboxy end of the glucosidase domain and the glycogen-binding domain. Agl knockout mice presented serious hepatomegaly, but we did not observe signs of cirrhosis or adenomas. In affected tissues, glycogen storage was higher than in wild-type mice, even in the central nervous system which has never been tested in GSDIII patients. The biochemical findings were in accordance with histological data, which clearly documented tissue impairment due to glycogen accumulation. Indeed, electron microscopy revealed the disruption of contractile units due to glycogen infiltrations. Furthermore, adult Agl knockout animals appeared less prompt to move, and they exhibited kyphosis. Three-mo-old Agl knockout mice could not run, and adult mice showed exercise intolerance. In addition, older affected animals exhibited an accelerated respiratory rate even at basal conditions. This observation was correlated with severe glycogen accumulation in the diaphragm. Diffuse glycogen deposition was observed in the tongues of affected mice. Our results demonstrate that this Agl knockout mouse is a reliable model for human glycogenosis type III, as it recapitulates the essential phenotypic features of the disease.
- Published
- 2014
40. The wide spectrum of clinical phenotypes of spinal muscular atrophy with respiratory distress type 1: A systematic review
- Author
-
Chiara Simone, Francesca Magri, Giulietta Riboldi, Irene Faravelli, Francesca Porro, Stefania Corti, Paola Rinchetti, Chiara Zanetta, and Monica Nizzardo
- Subjects
Respiratory Distress Syndrome, Newborn ,Pathology ,medicine.medical_specialty ,Palsy ,Respiratory distress ,business.industry ,Genetic heterogeneity ,Neuroimaging ,Spinal muscular atrophy ,medicine.disease ,Phenotype ,DNA-Binding Proteins ,Muscular Atrophy, Spinal ,Low birth weight ,Atrophy ,Autonomic Nervous System Diseases ,Neurology ,Mutation ,medicine ,Humans ,Neurology (clinical) ,medicine.symptom ,business ,Transcription Factors - Abstract
Spinal muscular atrophy with respiratory distress type 1 (SMARD1), also known as distal spinal–muscular atrophy 1 (DSMA10), is an autosomal recessive type of spinal muscular atrophy that is related to mutations in the IGHMBP2 gene, which encodes for the immunoglobulin μ-binding protein. SMARD1 patients usually present low birth weight, diaphragmatic palsy and distal muscular atrophy. Clinical features are still the most important factor that leads to the diagnosis of SMARD1, due to the fact that IGHMBP2 gene mutations are characterized by significant phenotypic heterogeneity. In the present review, we will systematically discuss the genetic, clinical and neuropathological features of SMARD1 in order to provide a complete overview of SMARD1 variable clinical presentations and of the most important diagnostic tools which can be used to identify and properly manage affected individuals. This background is crucial also in the perspective of the development of novel therapeutic strategies for this still orphan disorder.
- Published
- 2014
41. iPSC-Derived Neural Stem Cells Act via Kinase Inhibition to Exert Neuroprotective Effects in Spinal Muscular Atrophy with Respiratory Distress Type 1
- Author
-
Monica Nizzardo, Chiara Simone, Giulietta Riboldi, Margherita Ruggieri, Sabrina Salani, Stefania Corti, Giacomo P. Comi, Federica Rizzo, Monica Bucchia, and Nereo Bresolin
- Subjects
Induced Pluripotent Stem Cells ,Transplantation, Heterologous ,Kaplan-Meier Estimate ,Protein Serine-Threonine Kinases ,Biology ,Biochemistry ,Neuroprotection ,Article ,Muscular Atrophy, Spinal ,Glycogen Synthase Kinase 3 ,Mice ,Neural Stem Cells ,Neurotrophic factors ,Genetics ,medicine ,Animals ,Humans ,Cell Lineage ,Nerve Growth Factors ,Axon ,Induced pluripotent stem cell ,lcsh:QH301-705.5 ,Motor Neurons ,lcsh:R5-920 ,Respiratory Distress Syndrome, Newborn ,Cell Differentiation ,Cell Biology ,Spinal muscular atrophy ,Anatomy ,Motor neuron ,medicine.disease ,Axons ,Coculture Techniques ,Neural stem cell ,Transplantation ,Disease Models, Animal ,medicine.anatomical_structure ,lcsh:Biology (General) ,nervous system ,lcsh:Medicine (General) ,Neuroscience ,Developmental Biology - Abstract
Summary Spinal muscular atrophy with respiratory distress type 1 (SMARD1) is a motor neuron disease caused by mutations in the IGHMBP2 gene, without a cure. Here, we demonstrate that neural stem cells (NSCs) from human-induced pluripotent stem cells (iPSCs) have therapeutic potential in the context of SMARD1. We show that upon transplantation NSCs can appropriately engraft and differentiate in the spinal cord of SMARD1 animals, ameliorating their phenotype, by protecting their endogenous motor neurons. To evaluate the effect of NSCs in the context of human disease, we generated human SMARD1-iPSCs motor neurons that had a significantly reduced survival and axon length. Notably, the coculture with NSCs ameliorate these disease features, an effect attributable to the production of neurotrophic factors and their dual inhibition of GSK-3 and HGK kinases. Our data support the role of iPSC as SMARD1 disease model and their translational potential for therapies in motor neuron disorders., Graphical Abstract, Highlights • Transplantation of human NSCs ameliorates the SMARD1 phenotype in a disease model • NSCs improve the disease features of SMARD1 motor neurons inhibiting GSK-3 and HGK, Corti and colleagues show that neural stem cells (NSCs) from human-induced pluripotent stem cells (hiPSCs) have therapeutic potential for Spinal Muscular Atrophy with Respiratory Distress Type 1 SMARD1. Transplanted NSCs engraft in the spinal cord of SMARD1 animals, ameliorating their phenotype. SMARD1 hiPSC-derived motor neurons recapitulate disease features that are improved by NSC coculture through GSK-3 and HGK kinase-modulated pathways.
- Published
- 2014
- Full Text
- View/download PDF
42. Antisense Oligonucleotide Therapy for the Treatment of C9ORF72 ALS/FTD Diseases
- Author
-
Michela Ranieri, Stefania Corti, Nereo Bresolin, Giulietta Riboldi, Chiara Simone, Francesca Magri, Giacomo P. Comi, Monica Nizzardo, and Chiara Zanetta
- Subjects
Motor Neurons ,DNA Repeat Expansion ,C9orf72 Protein ,Oligonucleotide ,Amyotrophic Lateral Sclerosis ,Neuroscience (miscellaneous) ,Proteins ,RNA ,Oligonucleotides, Antisense ,Biology ,medicine.disease ,Cellular and Molecular Neuroscience ,Neurology ,C9orf72 ,Frontotemporal Dementia ,Antisense Technology ,medicine ,Humans ,Amyotrophic lateral sclerosis ,Trinucleotide repeat expansion ,Neuroscience ,Frontotemporal dementia - Abstract
Motor neuron disorders, and particularly amyotrophic lateral sclerosis (ALS), are fatal diseases that are due to the loss of motor neurons in the brain and spinal cord, with progressive paralysis and premature death. It has been recently shown that the most frequent genetic cause of ALS, frontotemporal dementia (FTD), and other neurological diseases is the expansion of a hexanucleotide repeat (GGGGCC) in the non-coding region of the C9ORF72 gene. The pathogenic mechanisms that produce cell death in the presence of this expansion are still unclear. One of the most likely hypotheses seems to be the gain-of-function that is achieved through the production of toxic RNA (able to sequester RNA-binding protein) and/or toxic proteins. In recent works, different authors have reported that antisense oligonucleotides complementary to the C9ORF72 RNA transcript sequence were able to significantly reduce RNA foci generated by the expanded RNA, in affected cells. Here, we summarize the recent findings that support the idea that the buildup of "toxic" RNA containing the GGGGCC repeat contributes to the death of motor neurons in ALS and also suggest that the use of antisense oligonucleotides targeting this transcript is a promising strategy for treating ALS/frontotemporal lobe dementia (FTLD) patients with the C9ORF72 repeat expansion. These data are particularly important, given the state of the art antisense technology, and they allow researchers to believe that a clinical application of these discoveries will be possible soon.
- Published
- 2014
43. Stem cell transplantation for amyotrophic lateral sclerosis: therapeutic potential and perspectives on clinical translation
- Author
-
Chiara Simone, Stefania Corti, Chiara Zanetta, Giacomo P. Comi, Giulietta Riboldi, Monica Nizzardo, Nereo Bresolin, and Irene Faravelli
- Subjects
Pathology ,medicine.medical_specialty ,Neurogenesis ,Induced Pluripotent Stem Cells ,Mesenchymal Stem Cell Transplantation ,Translational Research, Biomedical ,Cellular and Molecular Neuroscience ,Therapeutic approach ,Neural Stem Cells ,medicine ,Animals ,Humans ,Amyotrophic lateral sclerosis ,Induced pluripotent stem cell ,Molecular Biology ,Embryonic Stem Cells ,Injections, Spinal ,Motor Neurons ,Pharmacology ,Clinical Trials as Topic ,business.industry ,Therapies, Investigational ,Amyotrophic Lateral Sclerosis ,Cell Biology ,Motor neuron ,medicine.disease ,Neural stem cell ,Transplantation ,Disease Models, Animal ,medicine.anatomical_structure ,Cellular Microenvironment ,Spinal Cord ,Molecular Medicine ,Stem cell ,business ,Neuroglia ,Neuroscience ,Stem Cell Transplantation - Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal neurological disease characterized by degeneration of upper and lower motor neurons. There are currently no clinically impactful treatments for this disorder. Death occurs 3-5 years after diagnosis, usually due to respiratory failure. ALS pathogenesis seems to involve several pathological mechanisms (i.e., oxidative stress, inflammation, and loss of the glial neurotrophic support, glutamate toxicity) with different contributions from environmental and genetic factors. This multifaceted combination highlights the concept that an effective therapeutic approach should counteract simultaneously different aspects: stem cell therapies are able to maintain or rescue motor neuron function and modulate toxicity in the central nervous system (CNS) at the same time, eventually representing the most comprehensive therapeutic approach for ALS. To achieve an effective cell-mediated therapy suitable for clinical applications, several issues must be addressed, including the identification of the most performing cell source, a feasible administration protocol, and the definition of therapeutic mechanisms. The method of cell delivery represents a major issue in developing cell-mediated approaches since the cells, to be effective, need to be spread across the CNS, targeting both lower and upper motor neurons. On the other hand, there is the need to define a strategy that could provide a whole distribution without being too invasive or burdened by side effects. Here, we review the recent advances regarding the therapeutic potential of stem cells for ALS with a focus on the minimally invasive strategies that could facilitate an extensive translation to their clinical application.
- Published
- 2014
44. Molecular Therapeutic Strategies for Spinal Muscular Atrophies: Current and Future Clinical Trials
- Author
-
Chiara Zanetta, Giacomo P. Comi, Erika Monguzzi, Chiara Simone, Nereo Bresolin, Monica Nizzardo, and Stefania Corti
- Subjects
medicine.medical_specialty ,Genetic enhancement ,SMN1 ,Disease ,Bioinformatics ,Muscular Atrophy, Spinal ,chemistry.chemical_compound ,medicine ,Humans ,Pharmacology (medical) ,Molecular Targeted Therapy ,Pharmacology ,Clinical Trials as Topic ,business.industry ,Genetic Therapy ,Spinal muscular atrophy ,medicine.disease ,Spinal muscular atrophies ,SMA ,Survival of Motor Neuron 1 Protein ,Survival of Motor Neuron 2 Protein ,Clinical trial ,chemistry ,Physical therapy ,Olesoxime ,business - Abstract
Background Spinal muscular atrophy (SMA) is an autosomal recessive motor neuron disease caused by mutations in the survival motor neuron gene ( SMN1 ) and the leading genetic cause of infant mortality. Currently, there is no effective treatment other than supportive care. Objective This article provides a general overview of the main aspects that need to be taken into account to design a more efficient clinical trial and to summarize the most promising molecular trials that are currently in development or are being planned for the treatment of SMA. Methods A systematic review of the literature was performed, identifying key clinical trials involving novel molecular therapies in SMA. In addition, abstracts presented at the meetings of the Families of Spinal Muscular Atrophy were searched and the Families of Spinal Muscular Atrophy Web site was carefully analyzed. Finally, a selection of SMA clinical trials registered at clinical- trials.gov has been included in the article. Results The past decade has seen a marked advancement in the understanding of both SMA genetics and molecular mechanisms. New molecules targeting SMN have shown promise in preclinical studies, and various clinical trials have started to test the drugs that were discovered through basic research. Conclusions Both preclinical and early clinical trial results involving novel molecular therapies suggest that the clinical care paradigm in SMA will soon change.
- Published
- 2014
45. CSF transplantation of a specific iPSC-derived neural stem cell subpopulation ameliorates the disease phenotype in a mouse model of spinal muscular atrophy with respiratory distress type 1
- Author
-
Elena Trombetta, Giulia Forotti, Giacomo P. Comi, Agnese Ramirez, Stefano Gatti, Monica Nizzardo, Stefania Corti, Monica Bucchia, and Nereo Bresolin
- Subjects
0301 basic medicine ,Necroptosis ,Induced Pluripotent Stem Cells ,CXCR4 ,Muscular Atrophy, Spinal ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Neural Stem Cells ,Developmental Neuroscience ,medicine ,Animals ,Humans ,Induced pluripotent stem cell ,Respiratory Distress Syndrome, Newborn ,business.industry ,Spinal muscular atrophy ,Motor neuron ,medicine.disease ,Neural stem cell ,3. Good health ,Transplantation ,Disease Models, Animal ,Phenotype ,030104 developmental biology ,medicine.anatomical_structure ,Neurology ,Cancer research ,Stem cell ,business ,030217 neurology & neurosurgery ,Stem Cell Transplantation - Abstract
Spinal muscular atrophy with respiratory distress type 1 (SMARD1) is a genetic motor neuron disease affecting infants. This condition is caused by mutations in the IGHMBP2 gene and currently has no cure. Stem cell transplantation is a potential therapeutic strategy for motor neuron diseases such as SMARD1, exerting beneficial effects both by replacing cells and by providing support to endogenous motor neurons. In this work, we demonstrate that human induced pluripotent stem cell (iPSC)-derived neural stem cells (NSCs) selected for the expression of specific markers, namely, Lewis X, CXCR4 and beta 1 integrin, and pretreated with neurotrophic factors and apoptosis/necroptosis inhibitors were able to effectively migrate and engraft into the host parenchyma after administration into the cerebrospinal fluid in a SMARD1 mouse model. We were able to detect donor cells in the ventral horn of the spinal cord and observe improvements in neuropathological features, particularly preservation of the integrity of the motor unit, that were correlated with amelioration of the SMARD1 disease phenotype in terms of neuromuscular function and lifespan. This minimally invasive stem cell approach can confer major advantages in the context of cell-mediated therapy for patients with neurodegenerative diseases.
- Published
- 2019
46. In vitro neurogenesis: development and functional implications of iPSC technology
- Author
-
Enrico Bertini, Ginevra Zanni, Stefania Corti, Claudia Compagnucci, and Monica Nizzardo
- Subjects
Pharmacology ,Cell growth ,Neurogenesis ,Induced Pluripotent Stem Cells ,Cell ,Neurodegenerative Diseases ,Cell Biology ,Biology ,Neural stem cell ,In vitro ,Functional networks ,Cellular and Molecular Neuroscience ,medicine.anatomical_structure ,Neural Stem Cells ,medicine ,Animals ,Humans ,Molecular Medicine ,Human embryogenesis ,Induced pluripotent stem cell ,Molecular Biology ,Neuroscience ,Embryonic Stem Cells ,Stem Cell Transplantation - Abstract
Neurogenesis is the developmental process regulating cell proliferation of neural stem cells, determining their differentiation into glial and neuronal cells, and orchestrating their organization into finely regulated functional networks. Can this complex process be recapitulated in vitro using induced pluripotent stem cell (iPSC) technology? Can neurodevelopmental and neurodegenerative diseases be modeled using iPSCs? What is the potential of iPSC technology in neurobiology? What are the recent advances in the field of neurological diseases? Since the applications of iPSCs in neurobiology are based on the capacity to regulate in vitro differentiation of human iPSCs into different neuronal subtypes and glial cells, and the possibility of obtaining iPSC-derived neurons and glial cells is based on and hindered by our poor understanding of human embryonic development, we reviewed current knowledge on in vitro neural differentiation from a developmental and cellular biology perspective. We highlight the importance to further advance our understanding on the mechanisms controlling in vivo neurogenesis in order to efficiently guide neurogenesis in vitro for cell modeling and therapeutical applications of iPSCs technology.
- Published
- 2013
47. Minimally invasive transplantation of iPSC-derived ALDHhiSSCloVLA4+ neural stem cells effectively improves the phenotype of an amyotrophic lateral sclerosis model
- Author
-
Chiara Zanetta, Monica Nizzardo, Chiara Simone, Irene Faravelli, Giulietta Riboldi, Federica Rizzo, Stefania Corti, Sabrina Salani, Margherita Ruggieri, Nereo Bresolin, and Giacomo P. Comi
- Subjects
Population ,Induced Pluripotent Stem Cells ,TRPV Cation Channels ,Mice, Transgenic ,Biology ,Integrin alpha4beta1 ,Mice ,Superoxide Dismutase-1 ,Neural Stem Cells ,Neurotrophic factors ,Genetics ,medicine ,Animals ,Humans ,Amyotrophic lateral sclerosis ,education ,Induced pluripotent stem cell ,Molecular Biology ,Genetics (clinical) ,education.field_of_study ,Superoxide Dismutase ,Amyotrophic Lateral Sclerosis ,General Medicine ,Articles ,Aldehyde Dehydrogenase ,medicine.disease ,Neural stem cell ,Transplantation ,Mice, Inbred C57BL ,Disease Models, Animal ,Phenotype ,nervous system ,Astrocytes ,Immunology ,Cancer research ,Stem cell ,Reprogramming ,Stem Cell Transplantation - Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal neurological disease characterized by the degeneration of motor neurons. Currently, there is no effective therapy for ALS. Stem cell transplantation is a potential therapeutic strategy for ALS, and the reprogramming of adult somatic cells into induced pluripotent stem cells (iPSCs) represents a novel cell source. In this study, we isolated a specific neural stem cell (NSC) population from human iPSCs based on high aldehyde dehydrogenase activity, low side scatter and integrin VLA4 positivity. We assessed the therapeutic effects of these NSCs on the phenotype of ALS mice after intrathecal or intravenous injections. Transplanted NSCs migrated and engrafted into the central nervous system via both routes of injection. Compared with control ALS, treated ALS mice exhibited improved neuromuscular function and motor unit pathology and significantly increased life span, in particular with the systemic administration of NSCs (15%). These positive effects are linked to multiple mechanisms, including production of neurotrophic factors and reduction of micro- and macrogliosis. NSCs induced a decrease in astrocyte number through the activation of the vanilloid receptor TRPV1. We conclude that minimally invasive injections of iPSC-derived NSCs can exert a therapeutic effect in ALS. This study contributes to advancements in iPSC-mediated approaches for treating ALS and other neurodegenerative diseases.
- Published
- 2013
48. Direct Reprogramming of Adult Somatic Cells into other Lineages: Past Evidence and Future Perspectives
- Author
-
Marianna Falcone, Monica Nizzardo, Giacomo P. Comi, Nereo Bresolin, Giulietta Riboldi, Chiara Simone, and Stefania Corti
- Subjects
Adult ,Somatic cell ,Cell ,Biomedical Engineering ,lcsh:Medicine ,Biology ,Models, Biological ,Regenerative medicine ,medicine ,Animals ,Humans ,Cell Lineage ,Induced pluripotent stem cell ,Transcription factor ,Genetics ,Transplantation ,lcsh:R ,Transdifferentiation ,Cell Biology ,Cellular Reprogramming ,Cell biology ,medicine.anatomical_structure ,Gene Expression Regulation ,Cell Transdifferentiation ,Stem cell ,Reprogramming - Abstract
Direct reprogramming of an adult cell into another differentiated lineage—such as fibroblasts into neurons, cardiomyocytes, or blood cells—without passage through an undifferentiated pluripotent stage is a new area of research that has recently emerged alongside stem cell technology and induced pluripotent stem cell reprogramming; indeed, this avenue of investigation has begun to play a central role in basic biological research and regenerative medicine. Even though the field seems new, its origins go back to the 1980s when it was demonstrated that differentiated adult cells can be converted into another cell lineage through the overexpression of transcription factors, establishing mature cell plasticity. Here, we retrace transdifferentiation experiments from the discovery of master control genes to recent in vivo reprogramming of one somatic cell into another from the perspective of possible applications for the development of new therapeutic approaches for human diseases.
- Published
- 2013
49. iPSC-derived LewisX+CXCR4+β1-integrin+ neural stem cells improve the amyotrophic lateral sclerosis phenotype by preserving motor neurons and muscle innervation in human and rodent models
- Author
-
Monica Bucchia, Giacomo P. Comi, Nereo Bresolin, Elena Trombetta, Agnese Ramirez, Monica Nizzardo, and Stefania Corti
- Subjects
0301 basic medicine ,Induced Pluripotent Stem Cells ,Lewis X Antigen ,Mice, Transgenic ,Biology ,Microgliosis ,Neuromuscular junction ,Cell Line ,Receptors, Interleukin-8A ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Neural Stem Cells ,Genetics ,medicine ,Animals ,Humans ,Amyotrophic lateral sclerosis ,Induced pluripotent stem cell ,Muscle, Skeletal ,Molecular Biology ,Genetics (clinical) ,Motor Neurons ,Muscle Denervation ,Superoxide Dismutase ,Integrin beta1 ,Amyotrophic Lateral Sclerosis ,General Medicine ,medicine.disease ,Allografts ,Neural stem cell ,Transplantation ,Disease Models, Animal ,030104 developmental biology ,medicine.anatomical_structure ,Immunology ,Neuroscience ,030217 neurology & neurosurgery ,Reinnervation ,Stem Cell Transplantation - Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal incurable neurodegenerative disease characterized by progressive degeneration of motor neurons (MNs), leading to relentless muscle paralysis. In the early stage of the disease, MN loss and consequent muscle denervation are compensated by axonal sprouting and reinnervation by the remaining MNs, but this mechanism is insufficient in the long term. Here, we demonstrate that induced pluripotent stem cell-derived neural stem cells (NSCs), in particular the subpopulation positive for LewisX-CXCR4-β1-integrin, enhance neuronal survival and axonal growth of human ALS-derived MNs co-cultured with toxic ALS astrocytes, acting on both autonomous and non-autonomous ALS disease features. Transplantation of this NSC fraction into transgenic SOD1G93A ALS mice protects MNs in vivo, promoting their ability to maintain neuromuscular junction integrity, inducing novel axonal sprouting and reducing macro- and microgliosis. These effects result in a significant increase in survival and an improved neuromuscular phenotype in transplanted SOD1G93A mice. Our findings suggest that effective protection of MN functional innervation can be achieved by modulation of multiple dysregulated cellular and molecular pathways in both MNs and glial cells. These pathways must be considered in designing therapeutic strategies for ALS patients.
- Published
- 2016
50. Morpholino-mediated SOD1 reduction ameliorates an amyotrophic lateral sclerosis disease phenotype
- Author
-
Gianna Ulzi, Mafalda Rizzuti, Monica Bucchia, Giacomo P. Comi, Stefania Corti, Andreina Bordoni, Agnese Ramirez, Nereo Bresolin, Stefano Gatti, Federica Rizzo, Chiara Simone, and Monica Nizzardo
- Subjects
0301 basic medicine ,Protein Folding ,Pathology ,medicine.medical_specialty ,Morpholino ,Induced Pluripotent Stem Cells ,SOD1 ,Oligonucleotides ,Apoptosis ,Mice, Transgenic ,Biology ,Article ,Mice ,03 medical and health sciences ,Superoxide Dismutase-1 ,0302 clinical medicine ,medicine ,Animals ,Humans ,Gene silencing ,Gene Silencing ,Axon ,Amyotrophic lateral sclerosis ,Inflammation ,Motor Neurons ,Multidisciplinary ,Cell Death ,Amyotrophic Lateral Sclerosis ,Neurotoxicity ,Motor neuron ,medicine.disease ,Axons ,Astrogliosis ,Mice, Inbred C57BL ,Disease Models, Animal ,030104 developmental biology ,medicine.anatomical_structure ,Spinal Cord ,Disease Progression ,Cancer research ,030217 neurology & neurosurgery ,HeLa Cells - Abstract
Neurotoxicity due to the accumulation of mutant proteins is thought to drive pathogenesis in neurodegenerative diseases. Mutations in superoxide dismutase 1 (SOD1) are linked to familial amyotrophic lateral sclerosis (fALS); these mutations result in progressive motor neuron death through one or more acquired toxicities. Interestingly, SOD1 is not only responsible for fALS but may also play a significant role in sporadic ALS; therefore, SOD1 represents a promising therapeutic target. Here, we report slowed disease progression, improved neuromuscular function, and increased survival in an in vivo ALS model following therapeutic delivery of morpholino oligonucleotides (MOs) designed to reduce the synthesis of human SOD1. Neuropathological analysis demonstrated increased motor neuron and axon numbers and a remarkable reduction in astrogliosis and microgliosis. To test this strategy in a human model, we treated human fALS induced pluripotent stem cell (iPSC)-derived motor neurons with MOs; these cells exhibited increased survival and reduced expression of apoptotic markers. Our data demonstrated the efficacy of MO-mediated therapy in mouse and human ALS models, setting the stage for human clinical trials.
- Published
- 2016
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.