Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
NPJ Regen Med ; 8(1): 33, 2023 Jul 10.
Article in English | MEDLINE | ID: mdl-37429840

ABSTRACT

Neurogenesis, crucial for brain resilience, is reduced in Alzheimer's disease (AD) that induces astroglial reactivity at the expense of the pro-neurogenic potential, and restoring neurogenesis could counteract neurodegenerative pathology. However, the molecular mechanisms promoting pro-neurogenic astroglial fate despite AD pathology are unknown. In this study, we used APP/PS1dE9 mouse model and induced Nerve growth factor receptor (Ngfr) expression in the hippocampus. Ngfr, which promotes neurogenic fate of astroglia during the amyloid pathology-induced neuroregeneration in zebrafish brain, stimulated proliferative and neurogenic outcomes. Histological analyses of the changes in proliferation and neurogenesis, single-cell transcriptomics, spatial proteomics, and functional knockdown studies showed that the induced expression of Ngfr reduced the reactive astrocyte marker Lipocalin-2 (Lcn2), which we found was sufficient to reduce neurogenesis in astroglia. Anti-neurogenic effects of Lcn2 was mediated by Slc22a17, blockage of which recapitulated the pro-neurogenicity by Ngfr. Long-term Ngfr expression reduced amyloid plaques and Tau phosphorylation. Postmortem human AD hippocampi and 3D human astroglial cultures showed elevated LCN2 levels correlate with reactive gliosis and reduced neurogenesis. Comparing transcriptional changes in mouse, zebrafish, and human AD brains for cell intrinsic differential gene expression and weighted gene co-expression networks revealed common altered downstream effectors of NGFR signaling, such as PFKP, which can enhance proliferation and neurogenesis in vitro when blocked. Our study suggests that the reactive non-neurogenic astroglia in AD can be coaxed to a pro-neurogenic fate and AD pathology can be alleviated with Ngfr. We suggest that enhancing pro-neurogenic astroglial fate may have therapeutic ramifications in AD.

2.
Life (Basel) ; 13(6)2023 May 25.
Article in English | MEDLINE | ID: mdl-37374039

ABSTRACT

Motor neuron diseases (MNDs) are a heterogeneous group of disorders that affect the cranial and/or spinal motor neurons (spMNs), spinal sensory neurons and the muscular system. Although they have been investigated for decades, we still lack a comprehensive understanding of the underlying molecular mechanisms; and therefore, efficacious therapies are scarce. Model organisms and relatively simple two-dimensional cell culture systems have been instrumental in our current knowledge of neuromuscular disease pathology; however, in the recent years, human 3D in vitro models have transformed the disease-modeling landscape. While cerebral organoids have been pursued the most, interest in spinal cord organoids (SCOs) is now also increasing. Pluripotent stem cell (PSC)-based protocols to generate SpC-like structures, sometimes including the adjacent mesoderm and derived skeletal muscle, are constantly being refined and applied to study early human neuromuscular development and disease. In this review, we outline the evolution of human PSC-derived models for generating spMN and recapitulating SpC development. We also discuss how these models have been applied to exploring the basis of human neurodevelopmental and neurodegenerative diseases. Finally, we provide an overview of the main challenges to overcome in order to generate more physiologically relevant human SpC models and propose some exciting new perspectives.

3.
Front Cell Neurosci ; 16: 861202, 2022.
Article in English | MEDLINE | ID: mdl-35875350

ABSTRACT

Background: Protein aggregates are degraded via the autophagy-lysosome pathway and alterations in the lysosomal system leading to the accumulation of pathogenic proteins, including aggregates of α-synuclein in Parkinson's disease (PD). The importance of the endolysosomal transient receptor potential cation channel, mucolipin subfamily 1 (TRPML1) for the lysosomal function is highlighted by the fact that TRPML1 mutations cause the lysosomal storage disease mucolipidosis type IV. In this study, we investigated the mechanism by which activation of TRPML1 affects the degradation of α-synuclein. Methods: As a model of α-synuclein pathology, we expressed the pathogenic A53Tα-synuclein mutant in HEK293T cells. These cells were treated with the synthetic TRPML1 agonist ML-SA1. The amount of α-synuclein protein was determined by immunoblots. The abundance of aggregates and autolysosomal vesicles was determined by fluorescence microscopy and immunocytochemistry. Findings were confirmed by life-cell imaging and by application of ML-SA1 and the TRPML1 antagonist ML-SI3 to human dopaminergic neurons and human stem cell-derived neurons. Results: ML-SA1 reduced the percentage of HEK293T cells with α-synuclein aggregates and the amount of α-synuclein protein. The effect of ML-SA1 was blocked by pharmacological and genetic inhibition of autophagy. Consistent with TRPML function, it required the membrane lipid PI(3,5)P2, and cytosolic calcium. ML-SA1 shifted the composition of autophagosomes towards a higher fraction of mature autolysosomes, also in presence of α-synuclein. In neurons, inhibition of TRPML1 by its antagonist ML-SI3 blocked autophagosomal clearance, whereas the agonist ML-SA1 shifted the composition of a-synuclein particles towards a higher fraction of acidified particles. ML-SA1 was able to override the effect of Bafilomycin A1, which blocks the fusion of the autophagosome and lysosome and its acidification. Conclusion: These findings suggest, that activating TRPML1 with ML-SA1 facilitates clearance of α-synuclein aggregates primarily by affecting the late steps of the autophagy, i.e., by promoting autophagosome maturation. In agreement with recent work by others, our findings indicate that TRPML1 might constitute a plausible therapeutic target for PD, that warrants further validation in rodent models of α-synuclein pathology.

4.
Nat Commun ; 13(1): 422, 2022 01 20.
Article in English | MEDLINE | ID: mdl-35058465

ABSTRACT

Despite advances in nuclease-based genome editing technologies, correcting human disease-causing genomic inversions remains a challenge. Here, we describe the potential use of a recombinase-based system to correct the 140 kb inversion of the F8 gene frequently found in patients diagnosed with severe Hemophilia A. Employing substrate-linked directed molecular evolution, we develop a coupled heterodimeric recombinase system (RecF8) achieving 30% inversion of the target sequence in human tissue culture cells. Transient RecF8 treatment of endothelial cells, differentiated from patient-derived induced pluripotent stem cells (iPSCs) of a hemophilic donor, results in 12% correction of the inversion and restores Factor VIII mRNA expression. In this work, we present designer-recombinases as an efficient and specific means towards treatment of monogenic diseases caused by large gene inversions.


Subject(s)
Chromosome Inversion/genetics , Factor VIII/genetics , Recombinases/metabolism , Amino Acid Sequence , Base Sequence , Cell Differentiation , Clone Cells , Directed Molecular Evolution , Endothelial Cells/cytology , Endothelial Cells/metabolism , Exons/genetics , HEK293 Cells , HeLa Cells , Humans , Induced Pluripotent Stem Cells/metabolism , Inverted Repeat Sequences/genetics , Recombination, Genetic/genetics , Substrate Specificity , Whole Genome Sequencing
5.
Prog Mol Biol Transl Sci ; 172: 157-202, 2020.
Article in English | MEDLINE | ID: mdl-32620242

ABSTRACT

Motor neuron diseases (MNDs) are a wide group of neurodegenerative disorders characterized by the degeneration of a specific neuronal type located in the central nervous system, the motor neuron (MN). There are two main types of MNs, spinal and cortical MNs and depending on the type of MND, one or both types are affected. Cortical MNs innervate spinal MNs and these control a variety of cellular targets, being skeletal muscle their main one which is also affected in MNDs. A correct functionality of autophagy is necessary for the survival of all cellular types and it is particularly crucial for neurons, given their postmitotic and highly specialized nature. Numerous studies have identified alterations of autophagy activity in multiple MNDs. The scientific community has been particularly prolific in reporting the role that autophagy plays in the most common adult MND, amyotrophic lateral sclerosis, although many studies have started to identify physiological and pathological functions of this catabolic system in other MNDs, such as spinal muscular atrophy and spinal and bulbar muscular atrophy. The degradation of selective cargo by autophagy and how this process is altered upon the presence of MND-causing mutations is currently also a matter of intense investigation, particularly regarding the selective autophagic clearance of mitochondria. Thorough reviews on this field have been recently published. This chapter will cover the current knowledge on the functionality of autophagy and lysosomal homeostasis in the main MNDs and other autophagy-related topics in the MND field that have risen special interest in the research community.


Subject(s)
Autophagy , Motor Neuron Disease/pathology , Adult , Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/pathology , Animals , Autophagy/drug effects , Autophagy/physiology , Autophagy-Related Proteins/genetics , Autophagy-Related Proteins/physiology , C9orf72 Protein/deficiency , C9orf72 Protein/genetics , C9orf72 Protein/physiology , DNA Repeat Expansion , Disease Models, Animal , Endocytosis , Humans , Mice, Transgenic , Motor Neuron Disease/genetics , Muscular Atrophy, Spinal/genetics , Muscular Atrophy, Spinal/pathology , Mutation , Neurodegenerative Diseases/genetics , Neurodegenerative Diseases/pathology , Organelles , RNA-Binding Protein FUS/deficiency , RNA-Binding Protein FUS/genetics , RNA-Binding Protein FUS/physiology , TDP-43 Proteinopathies/genetics , TDP-43 Proteinopathies/pathology
6.
Cell Death Discov ; 6: 21, 2020.
Article in English | MEDLINE | ID: mdl-32337073

ABSTRACT

Glaucoma is a common age-related disease leading to progressive retinal ganglion cell (RGC) death, visual field defects and vision loss and is the second leading cause of blindness in the elderly worldwide. Mitochondrial dysfunction and impaired autophagy have been linked to glaucoma and induction of autophagy shows neuroprotective effects in glaucoma animal models. We have shown that autophagy decreases with aging in the retina and that autophagy can be neuroprotective for RGCs, but it is currently unknown how aging and autophagy deficiency impact RGCs susceptibility and survival. Using the optic nerve crush model in young and olWelcome@1234d Ambra1 +/gt (autophagy/beclin-1 regulator 1+/gt) mice we analysed the contribution of autophagy deficiency on retinal ganglion cell survival in an age dependent context. Interestingly, old Ambra1 +/gt mice showed decreased RGC survival after optic nerve crush in comparison to old Ambra1 +/+, an effect that was not observed in the young animals. Proteomics and mRNA expression data point towards altered oxidative stress response and mitochondrial alterations in old Ambra1 +/gt animals. This effect is intensified after RGC axonal damage, resulting in reduced oxidative stress response showing decreased levels of Nqo1, as well as failure of Nrf2 induction in the old Ambra1 +/gt. Old Ambra1 +/gt also failed to show increase in Bnip3l and Bnip3 expression after optic nerve crush, a response that is found in the Ambra1 +/+ controls. Primary RGCs derived from Ambra1 +/gt mice show decreased neurite projection and increased levels of apoptosis in comparison to Ambra1 +/+ animals. Our results lead to the conclusion that oxidative stress response pathways are altered in old Ambra1 +/gt mice leading to impaired damage responses upon additional external stress factors.

7.
Int J Mol Sci ; 21(5)2020 Mar 10.
Article in English | MEDLINE | ID: mdl-32164182

ABSTRACT

Mitochondrial damage plays a prominent role in glaucoma. The only way cells can degrade whole mitochondria is via autophagy, in a process called mitophagy. Thus, studying mitophagy in the context of glaucoma is essential to understand the disease. Up to date limited tools are available for analyzing mitophagy in vivo. We have taken advantage of the mito-QC reporter, a recently generated mouse model that allows an accurate mitophagy assessment to fill this gap. We used primary RGCs and retinal explants derived from mito-QC mice to quantify mitophagy activation in vitro and ex vivo. We also analyzed mitophagy in retinal ganglion cells (RGCs), in vivo, using different mitophagy inducers, as well as after optic nerve crush (ONC) in mice, a commonly used surgical procedure to model glaucoma. Using mito-QC reporter we quantified mitophagy induced by several known inducers in primary RGCs in vitro, ex vivo and in vivo. We also found that RGCs were rescued from some glaucoma relevant stress factors by incubation with the iron chelator deferiprone (DFP). Thus, the mito-QC reporter-based model is a valuable tool for accurately analyzing mitophagy in the context of glaucoma.


Subject(s)
Deferiprone/pharmacology , Genes, Reporter , Glaucoma/metabolism , Iron Chelating Agents/pharmacology , Mitochondria/metabolism , Retinal Ganglion Cells/cytology , Animals , Cell Survival/drug effects , Cells, Cultured , Disease Models, Animal , Glaucoma/etiology , Humans , Mice , Mitophagy , Primary Cell Culture , Rats , Retinal Ganglion Cells/drug effects , Retinal Ganglion Cells/metabolism
8.
J Clin Invest ; 128(7): 3008-3023, 2018 07 02.
Article in English | MEDLINE | ID: mdl-29672276

ABSTRACT

Spinal muscular atrophy (SMA), a degenerative motor neuron (MN) disease, caused by loss of functional survival of motor neuron (SMN) protein due to SMN1 gene mutations, is a leading cause of infant mortality. Increasing SMN levels ameliorates the disease phenotype and is unanimously accepted as a therapeutic approach for patients with SMA. The ubiquitin/proteasome system is known to regulate SMN protein levels; however, whether autophagy controls SMN levels remains poorly explored. Here, we show that SMN protein is degraded by autophagy. Pharmacological and genetic inhibition of autophagy increases SMN levels, while induction of autophagy decreases these levels. SMN degradation occurs via its interaction with the autophagy adapter p62 (also known as SQSTM1). We also show that SMA neurons display reduced autophagosome clearance, increased p62 and ubiquitinated proteins levels, and hyperactivated mTORC1 signaling. Importantly, reducing p62 levels markedly increases SMN and its binding partner gemin2, promotes MN survival, and extends lifespan in fly and mouse SMA models, revealing p62 as a potential new therapeutic target for the treatment of SMA.


Subject(s)
Muscular Atrophy, Spinal/drug therapy , Muscular Atrophy, Spinal/metabolism , SMN Complex Proteins/metabolism , Sequestosome-1 Protein/antagonists & inhibitors , Animals , Autophagy , Cells, Cultured , Disease Models, Animal , Gene Knockdown Techniques , HEK293 Cells , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Motor Neurons/metabolism , Muscular Atrophy, Spinal/pathology , Mutation , Phenotype , Proteolysis , RNA, Small Interfering/genetics , SMN Complex Proteins/deficiency , SMN Complex Proteins/genetics , Sequestosome-1 Protein/genetics , Sequestosome-1 Protein/metabolism , Survival of Motor Neuron 1 Protein/antagonists & inhibitors , Survival of Motor Neuron 1 Protein/genetics , Survival of Motor Neuron 1 Protein/metabolism , TOR Serine-Threonine Kinases/metabolism
9.
Development ; 145(4)2018 02 26.
Article in English | MEDLINE | ID: mdl-29483129

ABSTRACT

Autophagy is a catabolic pathway by which cellular components are delivered to the lysosome for degradation and recycling. Autophagy serves as a crucial intracellular quality control and repair mechanism but is also involved in cell remodelling during development and cell differentiation. In addition, mitophagy, the process by which damaged mitochondria undergo autophagy, has emerged as key regulator of cell metabolism. In recent years, a number of studies have revealed roles for autophagy and mitophagy in the regulation of stem cells, which represent the origin for all tissues during embryonic and postnatal development, and contribute to tissue homeostasis and repair throughout adult life. Here, we review these studies, focussing on the latest evidence that supports the quality control, remodelling and metabolic functions of autophagy during the activation, self-renewal and differentiation of embryonic, adult and cancer stem cells.


Subject(s)
Autophagy , Cell Differentiation/physiology , Cellular Reprogramming/physiology , Stem Cells/physiology , Animals , Humans
10.
Cells ; 6(4)2017 Oct 22.
Article in English | MEDLINE | ID: mdl-29065501

ABSTRACT

Autophagy is a catabolic pathway that mediates the degradation and recycling of intracellular components, and is a key player in a variety of physiological processes in cells and tissues. Recent studies of autophagy in the eye suggest that this pathway is fundamental for the preservation of retinal homeostasis. Given its accessible location outside the brain, the retina is an ideal organ in which to study the central nervous system and a wide range of neuronal processes, from development to neurodegeneration. Here we review several methods used to assess autophagy in the retina in both physiological and pathological conditions.

11.
Stem Cell Reports ; 9(2): 667-680, 2017 08 08.
Article in English | MEDLINE | ID: mdl-28712846

ABSTRACT

Amyotrophic lateral sclerosis (ALS) is a fatal and rapidly progressing motor neuron disease. Astrocytic factors are known to contribute to motor neuron degeneration and death in ALS. However, the role of astrocyte in promoting motor neuron protein aggregation, a disease hallmark of ALS, remains largely unclear. Here, using culture models of human motor neurons and primary astrocytes of different genotypes (wild-type or SOD1 mutant) and reactive states (non-reactive or reactive), we show that reactive astrocytes, regardless of their genotypes, reduce motor neuron health and lead to moderate neuronal loss. After prolonged co-cultures of up to 2 months, motor neurons show increased axonal and cytoplasmic protein inclusions characteristic of ALS. Reactive astrocytes induce protein aggregation in part by releasing transforming growth factor ß1 (TGF-ß1), which disrupts motor neuron autophagy through the mTOR pathway. These results reveal the important contribution of reactive astrocytes in promoting aspects of ALS pathology independent of genetic influences.


Subject(s)
Astrocytes/metabolism , Autophagy , Motor Neurons/metabolism , Protein Aggregation, Pathological , Transforming Growth Factor beta1/metabolism , Amyotrophic Lateral Sclerosis/etiology , Amyotrophic Lateral Sclerosis/metabolism , Amyotrophic Lateral Sclerosis/pathology , Animals , Astrocytes/pathology , Axons/metabolism , Cell Survival/genetics , Cells, Cultured , Cytoplasm/metabolism , Disease Models, Animal , Humans , Intermediate Filaments/metabolism , Mice , Mutation , Protein Aggregates/genetics , Signal Transduction , Superoxide Dismutase-1/genetics , Superoxide Dismutase-1/metabolism , TOR Serine-Threonine Kinases/metabolism
12.
J Med Chem ; 60(12): 4983-5001, 2017 06 22.
Article in English | MEDLINE | ID: mdl-28548834

ABSTRACT

Glycogen synthase kinase 3 ß (GSK-3ß) is a central target in several unmet diseases. To increase the specificity of GSK-3ß inhibitors in chronic treatments, we developed small molecules allowing subtle modulation of GSK-3ß activity. Design synthesis, structure-activity relationships, and binding mode of quinoline-3-carbohydrazide derivatives as allosteric modulators of GSK-3ß are presented here. Furthermore, we show how allosteric binders may overcome the ß-catenin side effects associated with strong GSK-3ß inhibition. The therapeutic potential of some of these modulators has been tested in human samples from patients with congenital myotonic dystrophy type 1 (CDM1) and spinal muscular atrophy (SMA) patients. We found that compound 53 improves delayed myogenesis in CDM1 myoblasts, while compounds 1 and 53 have neuroprotective properties in SMA-derived cells. These findings suggest that the allosteric modulators of GSK-3ß may be used for future development of drugs for DM1, SMA, and other chronic diseases where GSK-3ß inhibition exhibits therapeutic effects.


Subject(s)
Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Glycogen Synthase Kinase 3/antagonists & inhibitors , Allosteric Site , Chemistry Techniques, Synthetic , Drug Design , Drug Evaluation, Preclinical/methods , Glycogen Synthase Kinase 3/genetics , Glycogen Synthase Kinase 3/metabolism , Humans , Induced Pluripotent Stem Cells/drug effects , Molecular Docking Simulation , Molecular Dynamics Simulation , Muscular Atrophy, Spinal/drug therapy , Muscular Atrophy, Spinal/pathology , Myoblasts, Skeletal/drug effects , Myoblasts, Skeletal/pathology , Myotonic Dystrophy/drug therapy , Myotonic Dystrophy/pathology , Quinolines/chemistry , Quinolines/pharmacology , Structure-Activity Relationship , beta Catenin/metabolism
13.
Cell Rep ; 18(6): 1484-1498, 2017 02 07.
Article in English | MEDLINE | ID: mdl-28178525

ABSTRACT

The mechanism underlying selective motor neuron (MN) death remains an essential question in the MN disease field. The MN disease spinal muscular atrophy (SMA) is attributable to reduced levels of the ubiquitous protein SMN. Here, we report that SMN levels are widely variable in MNs within a single genetic background and that this heterogeneity is seen not only in SMA MNs but also in MNs derived from controls and amyotrophic lateral sclerosis (ALS) patients. Furthermore, cells with low SMN are more susceptible to cell death. These findings raise the important clinical implication that some SMN-elevating therapeutics might be effective in MN diseases besides SMA. Supporting this, we found that increasing SMN across all MN populations using an Nedd8-activating enzyme inhibitor promotes survival in both SMA and ALS-derived MNs. Altogether, our work demonstrates that examination of human neurons at the single-cell level can reveal alternative strategies to be explored in the treatment of degenerative diseases.


Subject(s)
Neuromuscular Diseases/metabolism , SMN Complex Proteins/metabolism , Amyotrophic Lateral Sclerosis/metabolism , Animals , Disease Models, Animal , Humans , Mice , Motor Neurons/metabolism , Muscular Atrophy, Spinal/metabolism , Single-Cell Analysis/methods , Spinal Cord/metabolism
14.
Cell Stem Cell ; 17(5): 569-84, 2015 Nov 05.
Article in English | MEDLINE | ID: mdl-26321202

ABSTRACT

Spinal muscular atrophy (SMA) is caused by mutations in the SMN1 gene. Because this gene is expressed ubiquitously, it remains poorly understood why motor neurons (MNs) are one of the most affected cell types. To address this question, we carried out RNA sequencing studies using fixed, antibody-labeled, and purified MNs produced from control and SMA patient-derived induced pluripotent stem cells (iPSCs). We found SMA-specific changes in MNs, including hyper-activation of the ER stress pathway. Functional studies demonstrated that inhibition of ER stress improves MN survival in vitro even in MNs expressing low SMN. In SMA mice, systemic delivery of an ER stress inhibitor that crosses the blood-brain barrier led to the preservation of spinal cord MNs. Therefore, our study implies that selective activation of ER stress underlies MN death in SMA. Moreover, the approach we have taken would be broadly applicable to the study of disease-prone human cells in heterogeneous cultures.


Subject(s)
Endoplasmic Reticulum Stress , Motor Neurons/metabolism , Muscular Atrophy, Spinal/metabolism , RNA/genetics , Sequence Analysis, RNA , Animals , Cell Death , Cell Differentiation , Cells, Cultured , Humans , Induced Pluripotent Stem Cells/metabolism , Induced Pluripotent Stem Cells/pathology , Mice , Mice, Inbred Strains , Mice, Transgenic , Motor Neurons/pathology , Muscular Atrophy, Spinal/pathology
15.
Aging Cell ; 12(3): 478-88, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23521856

ABSTRACT

Aging contributes to the appearance of several retinopathies and is the largest risk factor for aged-related macular degeneration, major cause of blindness in the elderly population. Accumulation of undegraded material as lipofuscin represents a hallmark in many pathologies of the aged eye. Autophagy is a highly conserved intracellular degradative pathway that plays a critical role in the removal of damaged cell components to maintain the cellular homeostasis. A decrease in autophagic activity with age observed in many tissues has been proposed to contribute to the aggravation of age-related diseases. However, the participation of different autophagic pathways to the retina physiopathology remains unknown. Here, we describe a marked reduction in macroautophagic activity in the retina with age, which coincides with an increase in chaperone-mediated autophagy (CMA). This increase in CMA is also observed during retinal neurodegeneration in the Atg5(flox/flox) ; nestin-Cre mice, a mouse model with downregulation of macroautophagy in neuronal precursors. In contrast to other cell types, this autophagic cross talk in retinal cells is not bi-directional and CMA inhibition renders cone photoreceptor very sensitive to stress. Temporal and cell-type-specific differences in the balance between autophagic pathways may be responsible for the specific pattern of visual loss that occurs with aging. Our results show for the first time a cross talk of different lysosomal proteolytic systems in the retina during normal aging and may help the development of new therapeutic intervention for age-dependent retinal diseases.


Subject(s)
Apoptosis , Autophagy , Macular Degeneration/physiopathology , Retina/physiology , Aging , Animals , Cell Line , Cellular Senescence , Female , Homeostasis , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Retina/physiopathology
16.
Autophagy ; 8(2): 286-8, 2012 Feb 01.
Article in English | MEDLINE | ID: mdl-22301991

ABSTRACT

In recent years autophagy modulation has been shown to reduce or increase neuronal cell death in several models of neurodegeneration. How autophagy exerts these dual effects is currently unknown. Here we review recent evidence from our laboratory demonstrating that autophagy can protect the cell soma after axonal traumatic injury. Damage in the optic nerve induces retinal ganglion cell (RGC) death in glaucoma and other retinal diseases and is often modeled by axotomy of the optic nerve in laboratory animals. Using this well-known model of RGC degeneration we show that autophagy is strongly upregulated following the insult and before cell death. Enhancement of autophagy by pharmacological treatment with rapamycin decreases the number of degenerating neurons. Conversely, axotomy in Atg4B (-/-) mice increases the number of dying cells in the retinal ganglion cell layer. Similar findings were observed in Atg5 (flox/flox) mice following specific downregulation of the autophagy regulator ATG5 in RGCs, by intravitreal injection of a cre-expressing vector. Taken together, these findings point to a cytoprotective role of autophagy following axonal damage in vivo.


Subject(s)
Autophagy , Axons/pathology , Animals , Axons/metabolism , Axotomy , Cell Count , Cell Survival , Down-Regulation , Humans , Mice , Models, Biological , Optic Nerve/pathology , Optic Nerve/surgery , Retinal Ganglion Cells/metabolism , Retinal Ganglion Cells/pathology , Up-Regulation
18.
J Neurosci ; 30(37): 12535-44, 2010 Sep 15.
Article in English | MEDLINE | ID: mdl-20844148

ABSTRACT

Mounting evidence suggests a role for autophagy dysregulation in Parkinson's disease (PD). The bulk degradation of cytoplasmic proteins (including α-synuclein) and organelles (such as mitochondria) is mediated by macroautophagy, which involves the sequestration of cytosolic components into autophagosomes (AP) and its delivery to lysosomes. Accumulation of AP occurs in postmortem brain samples from PD patients, which has been widely attributed to an induction of autophagy. However, the cause and pathogenic significance of these changes remain unknown. Here we found in the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine mouse model of PD that AP accumulation and dopaminergic cell death are preceded by a marked decrease in the amount of lysosomes within dopaminergic neurons. Lysosomal depletion was secondary to the abnormal permeabilization of lysosomal membranes induced by increased mitochondrial-derived reactive oxygen species. Lysosomal permeabilization resulted in a defective clearance and subsequent accumulation of undegraded AP and contributed directly to neurodegeneration by the ectopic release of lysosomal proteases into the cytosol. Lysosomal breakdown and AP accumulation also occurred in PD brain samples, where Lewy bodies were strongly immunoreactive for AP markers. Induction of lysosomal biogenesis by genetic or pharmacological activation of lysosomal transcription factor EB restored lysosomal levels, increased AP clearance and attenuated 1-methyl-4-phenylpyridinium-induced cell death. Similarly, the autophagy-enhancer compound rapamycin attenuated PD-related dopaminergic neurodegeneration, both in vitro and in vivo, by restoring lysosomal levels. Our results indicate that AP accumulation in PD results from defective lysosomal-mediated AP clearance secondary to lysosomal depletion. Restoration of lysosomal levels and function may thus represent a novel neuroprotective strategy in PD.


Subject(s)
Autophagy/physiology , Lysosomes/metabolism , Parkinsonian Disorders/metabolism , Parkinsonian Disorders/pathology , Aged , Animals , Animals, Newborn , Cell Death/physiology , Cell Line, Tumor , Cell Membrane Permeability/physiology , Cells, Cultured , Cytosol/enzymology , Cytosol/pathology , Disease Models, Animal , Dopamine/physiology , Humans , Lysosomes/pathology , Lysosomes/ultrastructure , Mice , Neurons/metabolism , Neurons/pathology , Neurons/ultrastructure , Peptide Hydrolases/metabolism , Phagosomes/metabolism , Phagosomes/pathology , Phagosomes/ultrastructure , Rats , Substantia Nigra/metabolism , Substantia Nigra/pathology , Substantia Nigra/ultrastructure
19.
Invest Ophthalmol Vis Sci ; 49(9): 4188-94, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18515565

ABSTRACT

PURPOSE: Retinitis pigmentosa (RP) is a heterogeneous group of inherited conditions that lead to blindness and for which there is no effective therapy. Apoptosis of photoreceptors is a common feature in animal models of the disease. Thus, the authors studied the therapeutic potential of proinsulin, an antiapoptotic molecule active during retinal development. METHODS: Transgenic mice expressing human proinsulin (hPi) in the skeletal muscle were generated in a mixed C57BL/6:SJL background and were back-crossed to a C57BL/6 background. Two independent lineages of transgenic mice were established in which hPi production in muscle was constitutive and not regulated by glucose levels. hPi levels in serum, muscle, and retina were determined with a commercial ELISA kit, visual function was evaluated by electroretinographic (ERG) recording, and programmed cell death was assessed by TUNEL. Immunohistochemistry was used to evaluate retinal structure preservation and oxidative damage. RESULTS: Transgenic expression of hPi in the rd10 retinal degeneration mouse model led to prolonged vision, as determined by ERG recording, in a manner that was related to the level of transgene expression. This attenuation of visual deterioration was correlated with a delay in photoreceptor apoptosis and with the preservation of retinal cytoarchitecture, particularly that of the cones. CONCLUSIONS: These results provide a new basis for possible therapies to counteract retinitis pigmentosa and a new tool to characterize the mechanisms involved in the progress of retinal neurodegeneration.


Subject(s)
Apoptosis , Proinsulin/toxicity , Retinal Degeneration/chemically induced , Retinitis Pigmentosa/physiopathology , Vision Disorders/chemically induced , Animals , Apoptosis/drug effects , Crosses, Genetic , Deoxycytosine Nucleotides/metabolism , Disease Models, Animal , Electroretinography , Genotype , Humans , Mice , Mice, Inbred C57BL , Mice, Inbred Strains , Mice, Transgenic , Retinal Degeneration/pathology , Retinitis Pigmentosa/chemically induced , Retinitis Pigmentosa/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...