Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 42
Filter
1.
Hum Mol Genet ; 32(11): 1888-1900, 2023 05 18.
Article in English | MEDLINE | ID: mdl-36752535

ABSTRACT

Bi-allelic mutations in GBA1, the gene that encodes ß-glucocerebrosidase (GCase), cause Gaucher disease (GD), whereas mono-allelic mutations do not cause overt pathology. Yet mono- or bi-allelic GBA1 mutations are the highest known risk factor for Parkinson's disease (PD). GCase deficiency results in the accumulation of glucosylceramide (GluCer) and its deacylated metabolite glucosylsphingosine (GluSph). Brains from patients with neuronopathic GD have high levels of GluSph, and elevation of this lipid in GBA1-associated PD has been reported. To uncover the mechanisms involved in GBA1-associated PD, we used human induced pluripotent stem cell-derived dopaminergic (DA) neurons from patients harboring heterozygote mutations in GBA1 (GBA1/PD-DA neurons). We found that compared with gene-edited isogenic controls, GBA1/PD-DA neurons exhibit mammalian target of rapamycin complex 1 (mTORC1) hyperactivity, a block in autophagy, an increase in the levels of phosphorylated α-synuclein (129) and α-synuclein aggregation. These alterations were prevented by incubation with mTOR inhibitors. Inhibition of acid ceramidase, the lysosomal enzyme that deacylates GluCer to GluSph, prevented mTOR hyperactivity, restored autophagic flux and lowered α-synuclein levels, suggesting that GluSph was responsible for these alterations. Incubation of gene-edited wild type (WT) controls with exogenous GluSph recapitulated the mTOR/α-synuclein abnormalities of GBA1/PD neurons, and these phenotypic alterations were prevented when GluSph treatment was in the presence of mTOR inhibitors. We conclude that GluSph causes an aberrant activation of mTORC1, suppressing normal lysosomal functions, including the clearance of pathogenic α-synuclein species. Our results implicate acid ceramidase in the pathogenesis of GBA1-associated PD, suggesting that this enzyme is a potential therapeutic target for treating synucleinopathies caused by GCase deficiency.


Subject(s)
Gaucher Disease , Induced Pluripotent Stem Cells , Parkinson Disease , Humans , Parkinson Disease/metabolism , alpha-Synuclein/genetics , alpha-Synuclein/metabolism , Induced Pluripotent Stem Cells/metabolism , MTOR Inhibitors , Acid Ceramidase/genetics , Acid Ceramidase/metabolism , Glucosylceramidase/genetics , Glucosylceramidase/metabolism , Gaucher Disease/metabolism , Dopaminergic Neurons/metabolism , TOR Serine-Threonine Kinases/genetics , Mechanistic Target of Rapamycin Complex 1/genetics , Mutation , Lysosomes/metabolism
2.
J Med Virol ; 96(2): e29455, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38323709

ABSTRACT

Severe acute respiratory coronavirus 2 (SARS-CoV-2) causes neurological disease in the peripheral and central nervous system (PNS and CNS, respectively) of some patients. It is not clear whether SARS-CoV-2 infection or the subsequent immune response are the key factors that cause neurological disease. Here, we addressed this question by infecting human induced pluripotent stem cell-derived CNS and PNS neurons with SARS-CoV-2. SARS-CoV-2 infected a low number of CNS neurons and did not elicit a robust innate immune response. On the contrary, SARS-CoV-2 infected a higher number of PNS neurons. This resulted in expression of interferon (IFN) λ1, several IFN-stimulated genes and proinflammatory cytokines. The PNS neurons also displayed alterations characteristic of neuronal damage, as increased levels of sterile alpha and Toll/interleukin receptor motif-containing protein 1, amyloid precursor protein and α-synuclein, and lower levels of cytoskeletal proteins. Interestingly, blockade of the Janus kinase and signal transducer and activator of transcription pathway by Ruxolitinib did not increase SARS-CoV-2 infection, but reduced neuronal damage, suggesting that an exacerbated neuronal innate immune response contributes to pathogenesis in the PNS. Our results provide a basis to study coronavirus disease 2019 (COVID-19) related neuronal pathology and to test future preventive or therapeutic strategies.


Subject(s)
COVID-19 , Induced Pluripotent Stem Cells , Humans , SARS-CoV-2 , Immunity, Innate , Neurons
3.
Mol Psychiatry ; 26(10): 5733-5750, 2021 10.
Article in English | MEDLINE | ID: mdl-32632204

ABSTRACT

Mutations in pitrilysin metallopeptidase 1 (PITRM1), a mitochondrial protease involved in mitochondrial precursor processing and degradation, result in a slow-progressing syndrome characterized by cerebellar ataxia, psychotic episodes, and obsessive behavior, as well as cognitive decline. To investigate the pathogenetic mechanisms of mitochondrial presequence processing, we employed cortical neurons and cerebral organoids generated from PITRM1-knockout human induced pluripotent stem cells (iPSCs). PITRM1 deficiency strongly induced mitochondrial unfolded protein response (UPRmt) and enhanced mitochondrial clearance in iPSC-derived neurons. Furthermore, we observed increased levels of amyloid precursor protein and amyloid ß in PITRM1-knockout neurons. However, neither cell death nor protein aggregates were observed in 2D iPSC-derived cortical neuronal cultures. On the other hand, over time, cerebral organoids generated from PITRM1-knockout iPSCs spontaneously developed pathological features of Alzheimer's disease (AD), including the accumulation of protein aggregates, tau pathology, and neuronal cell death. Single-cell RNA sequencing revealed a perturbation of mitochondrial function in all cell types in PITRM1-knockout cerebral organoids, whereas immune transcriptional signatures were substantially dysregulated in astrocytes. Importantly, we provide evidence of a protective role of UPRmt and mitochondrial clearance against impaired mitochondrial presequence processing and proteotoxic stress. Here, we propose a novel concept of PITRM1-linked neurological syndrome whereby defects of mitochondrial presequence processing induce an early activation of UPRmt that, in turn, modulates cytosolic quality control pathways. Thus, our work supports a mechanistic link between mitochondrial function and common neurodegenerative proteinopathies.


Subject(s)
Alzheimer Disease , Induced Pluripotent Stem Cells , Alzheimer Disease/genetics , Amyloid beta-Peptides , Humans , Metalloendopeptidases , Mitochondria , Organoids
4.
Neurobiol Dis ; 127: 1-12, 2019 07.
Article in English | MEDLINE | ID: mdl-30711484

ABSTRACT

While the link between GBA and Parkinson's disease (PD) was initially unexpected, it is now well established that GBA mutations are the most frequent genetic risk for PD. GBA has also been linked to sporadic PD, dementia with Lewy bodies, and ageing. Thus, GBA represents a promising target to counteract brain disease and the age-related decline of lysosomal function. The exact mechanisms involved in the risk of developing PD in GBA mutation carriers are still unclear and research in this field has faced the major challenge of a lack of proper modeling systems. Induced pluripotent stem cells (iPSCs) as well as advances in disease modeling and genome editing have facilitated studies of human brain disease. With regard to GBA-PD, iPSCs offer several advantages including the possibility of investigating sphingolipid (SPL) biology in relevant cells, the role of dopamine metabolism as well as non-cell autonomous mechanisms that are likely involved in the disease process. This review will summarize findings that emerged from iPSC-based studies in the context of GBA-PD pathology and therapy. We also highlight current advantages and challenges of stem cell models for neurological disease modeling and drug discovery.


Subject(s)
Brain/pathology , Glucosylceramidase/genetics , Induced Pluripotent Stem Cells , Parkinson Disease/genetics , Animals , Gene Editing , Humans , Parkinson Disease/drug therapy , Parkinson Disease/pathology
5.
Trends Immunol ; 37(11): 719-721, 2016 11.
Article in English | MEDLINE | ID: mdl-27638127

ABSTRACT

Mitochondrial antigens can be presented by MHC molecules and initiate adaptive immune responses but the mechanisms of mitochondrial antigen presentation (MitAP) have remained mostly unknown. A recent study proposes a new model whereby MitAP is mediated by a vesicle transport pathway that is suppressed by the Parkinson's disease (PD) associated proteins PTEN-induced putative kinase 1 (PINK1) and Parkin. This discovery brings a new perspective on the link between mitochondrial dysfunction and autoimmunity in PD.


Subject(s)
Antigen Presentation , Autoantigens/metabolism , Mitochondria/metabolism , Parkinson Disease/immunology , Protein Kinases/metabolism , Secretory Vesicles/metabolism , Ubiquitin-Protein Ligases/metabolism , Adaptive Immunity , Animals , Autoantigens/immunology , Autoimmunity , Histocompatibility Antigens/metabolism , Humans , Mitochondria/immunology , Models, Immunological , Protein Transport , Vacuoles/metabolism
6.
Biochem Biophys Res Commun ; 473(3): 665-74, 2016 05 06.
Article in English | MEDLINE | ID: mdl-26930470

ABSTRACT

Recent progress in human pluripotent stem cell (hPSC) and genome editing technologies has opened up new avenues for the investigation of human biology in health and disease as well as the development of therapeutic applications. Gene editing approaches with programmable nucleases have been successfully established in hPSCs and applied to study gene function, develop novel animal models and perform genetic and chemical screens. Several studies now show the successful editing of disease-linked alleles in somatic and patient-derived induced pluripotent stem cells (iPSCs) as well as in animal models. Importantly, initial clinical trials have shown the safety of programmable nucleases for ex vivo somatic gene therapy. In this context, the unlimited proliferation potential and the pluripotent properties of iPSCs may offer advantages for gene targeting approaches. However, many technical and safety issues still need to be addressed before genome-edited iPSCs are translated into the clinical setting. Here, we provide an overview of the available genome editing systems and discuss opportunities and perspectives for their application in basic research and clinical practice, with a particular focus on hPSC based research and gene therapy approaches. Finally, we discuss recent research on human germline genome editing and its social and ethical implications.


Subject(s)
Gene Editing , Pluripotent Stem Cells/cytology , Alleles , Animals , Cell Nucleus/metabolism , Deoxyribonucleases/metabolism , Genetic Therapy/methods , Genome, Human , Homologous Recombination , Humans , Induced Pluripotent Stem Cells/cytology , Mutation
7.
Cell Mol Life Sci ; 70(22): 4259-73, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23665870

ABSTRACT

The etiology of Parkinson's disease (PD) is complex and most likely involves numerous environmental and heritable risk factors. Interestingly, many genetic variants, which have been linked to familial forms of PD or identified as strong risk factors, also play a critical role in modulating inflammatory responses. There has been considerable debate in the field as to whether inflammation is a driving force in neurodegeneration or simply represents a response to neuronal death. One emerging hypothesis is that inflammation plays a critical role in the early phases of neurodegeneration. In this review, we will discuss emerging aspects of both innate and adaptive immunity in the context of the pathogenesis of PD. We will highlight recent data from genetic and functional studies that strongly support the theory that genetic susceptibility plays an important role in modulating immune pathways and inflammatory reactions, which may precede and initiate neuronal dysfunction and subsequent neurodegeneration. A detailed understanding of such cellular and molecular inflammatory pathways is crucial to uncover pathogenic mechanisms linking sporadic and hereditary PD and devise tailored neuroprotective interventions.


Subject(s)
Inflammation/metabolism , Parkinson Disease/etiology , Animals , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Disease Models, Animal , Humans , Immune System/metabolism , Inflammation/genetics , Metabolic Networks and Pathways , Parkinson Disease/genetics , Parkinson Disease/prevention & control , Polymorphism, Genetic , Risk Factors
8.
J Neurosci ; 32(6): 2062-85, 2012 Feb 08.
Article in English | MEDLINE | ID: mdl-22323720

ABSTRACT

In Parkinson's disease (PD), neurogenesis is impaired in the subventricular zone (SVZ) of postmortem human PD brains, in primate nonhuman and rodent models of PD. The vital role of Wingless-type MMTV integration site (Wnt)/ß-catenin signaling in the modulation of neurogenesis, neuroprotection, and synaptic plasticity coupled to our recent findings uncovering an active role for inflammation and Wnt/ß-catenin signaling in MPTP-induced loss and repair of nigrostriatal dopaminergic (DAergic) neurons prompted us to study the impact of neuroinflammation and the Wnt/ß-catenin pathway in the response of SVZ neuroprogenitors (NPCs) in MPTP-treated mice. In vivo experiments, using bromodeoxyuridine and cell-specific markers, and ex vivo time course analyses documented an inverse correlation between the reduced proliferation of NPCs and the generation of new neuroblasts with the phase of maximal exacerbation of microglia reaction, whereas a shift in the microglia proinflammatory phenotype correlated with a progressive NPC recovery. Ex vivo and in vitro experiments using microglia-NPC coculture paradigms pointed to NADPH-oxidase (gpPHOX(91)), a major source of microglial ROS, and reactive nitrogen species as candidate inhibitors of NPC neurogenic potential via the activation of glycogen synthase 3 (pGSK-3ß(Tyr216)), leading to loss of ß-catenin, a chief downstream transcriptional effector. Accordingly, MPTP/MPP(+) (1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine) caused ß-catenin downregulation and pGSK-3ß(Tyr216) overexpression, whereas manipulation of Wnt/ß-catenin signaling with RNA interference-mediated GSK-3ß knockdown or GSK-3ß antagonism reversed MPTP-induced neurogenic impairment ex vivo/in vitro or in vivo. Reciprocally, pharmacological modulation of inflammation prevented ß-catenin downregulation and restored neurogenesis, suggesting the possibility to modulate this endogenous system with potential consequences for DAergic neuroprotection and self-repair.


Subject(s)
Inflammation Mediators/administration & dosage , MPTP Poisoning/metabolism , Neuronal Plasticity/physiology , Parkinson Disease/metabolism , Receptor Cross-Talk/physiology , Stem Cells/metabolism , Wnt Signaling Pathway/physiology , beta Catenin/antagonists & inhibitors , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/administration & dosage , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/therapeutic use , Animals , Cells, Cultured , Coculture Techniques , Disease Models, Animal , Gene Knockdown Techniques/methods , Inflammation Mediators/physiology , MPTP Poisoning/drug therapy , MPTP Poisoning/pathology , Male , Mice , Mice, Inbred C57BL , Neurogenesis/drug effects , Neurogenesis/physiology , Neuronal Plasticity/drug effects , Neurons/drug effects , Neurons/metabolism , Neurons/pathology , Neuroprotective Agents/administration & dosage , Neuroprotective Agents/therapeutic use , Parkinson Disease/drug therapy , Parkinson Disease/pathology , Receptor Cross-Talk/drug effects , Stem Cells/drug effects , Stem Cells/pathology , Wnt Signaling Pathway/drug effects , beta Catenin/metabolism
9.
Proc Natl Acad Sci U S A ; 107(36): 15921-6, 2010 Sep 07.
Article in English | MEDLINE | ID: mdl-20798034

ABSTRACT

Recent advances in deriving induced pluripotent stem (iPS) cells from patients offer new possibilities for biomedical research and clinical applications, as these cells could be used for autologous transplantation. We differentiated iPS cells from patients with Parkinson's disease (PD) into dopaminergic (DA) neurons and show that these DA neurons can be transplanted without signs of neurodegeneration into the adult rodent striatum. The PD patient iPS (PDiPS) cell-derived DA neurons survived at high numbers, showed arborization, and mediated functional effects in an animal model of PD as determined by reduction of amphetamine- and apomorphine-induced rotational asymmetry, but only a few DA neurons projected into the host striatum at 16 wk after transplantation. We next applied FACS for the neural cell adhesion molecule NCAM on differentiated PDiPS cells before transplantation, which resulted in surviving DA neurons with functional effects on amphetamine-induced rotational asymmetry in a 6-OHDA animal model of PD. Morphologically, we found that PDiPS cell-derived non-DA neurons send axons along white matter tracts into specific close and remote gray matter target areas in the adult brain. Such findings establish the transplantation of human PDiPS cell-derived neurons as a long-term in vivo method to analyze potential disease-related changes in a physiological context. Our data also demonstrate proof of principle of survival and functional effects of PDiPS cell-derived DA neurons in an animal model of PD and encourage further development of differentiation protocols to enhance growth and function of implanted PDiPS cell-derived DA neurons in regard to potential therapeutic applications.


Subject(s)
Parkinson Disease/surgery , Pluripotent Stem Cells/cytology , Animals , Humans , Parkinson Disease/pathology , Rats
10.
FEBS Open Bio ; 13(9): 1558-1579, 2023 09.
Article in English | MEDLINE | ID: mdl-37219461

ABSTRACT

Glycolipid balance is key to normal body function, and its alteration can lead to a variety of diseases involving multiple organs and tissues. Glycolipid disturbances are also involved in Parkinson's disease (PD) pathogenesis and aging. Increasing evidence suggests that glycolipids affect cellular functions beyond the brain, including the peripheral immune system, intestinal barrier, and immunity. Hence, the interplay between aging, genetic predisposition, and environmental exposures could initiate systemic and local glycolipid changes that lead to inflammatory reactions and neuronal dysfunction. In this review, we discuss recent advances in the link between glycolipid metabolism and immune function and how these metabolic changes can exacerbate immunological contributions to neurodegenerative diseases, with a focus on PD. Further understanding of the cellular and molecular mechanisms that control glycolipid pathways and their impact on both peripheral tissues and the brain will help unravel how glycolipids shape immune and nervous system communication and the development of novel drugs to prevent PD and promote healthy aging.


Subject(s)
Neurodegenerative Diseases , Parkinson Disease , Humans , Parkinson Disease/genetics , Glycolipids , Inflammation , Brain/pathology
11.
Cell Rep ; 42(5): 112420, 2023 05 30.
Article in English | MEDLINE | ID: mdl-37083331

ABSTRACT

Sun et al. demonstrate that defects in autophagy cause nicotinamide adenine dinucleotide (NAD) depletion and neurotoxicity.1 Restoring NAD levels rescues cytotoxicity in autophagy-deficient neurons, providing a potential therapy for neurodegenerative and lysosomal storage diseases associated with autophagy defects.


Subject(s)
Brain Diseases , NAD , Humans , Neurons , Autophagy
12.
Cells ; 12(21)2023 10 31.
Article in English | MEDLINE | ID: mdl-37947628

ABSTRACT

Parkinson's disease (PD) is the second most prevalent neurodegenerative disease, characterized by the loss of midbrain dopaminergic neurons which leads to impaired motor and cognitive functions. PD is predominantly an idiopathic disease; however, about 5% of cases are linked to hereditary mutations. The most common mutation in both familial and sporadic PD is the G2019S mutation of leucine-rich repeat kinase 2 (LRRK2). Currently, it is not fully understood how this mutation leads to PD pathology. In this study, we isolated self-renewable, multipotent neural stem cells (NSCs) from induced pluripotent stem cells (iPSCs) harboring the G2019S LRRK2 mutation and compared them with their isogenic gene corrected counterparts using single-cell RNA-sequencing. Unbiased single-cell transcriptomic analysis revealed perturbations in many canonical pathways, specifically NRF2-mediated oxidative stress response, and glutathione redox reactions. Through various functional assays, we observed that G2019S iPSCs and NSCs exhibit increased basal levels of reactive oxygen species (ROS). We demonstrated that mutant cells show significant increase in the expression for KEAP1 and decrease in NRF2 associated with a reduced antioxidant response. The decreased viability of mutant NSCs in the H2O2-induced oxidative stress assay was rescued by two potent antioxidant drugs, PrC-210 at concentrations of 500 µM and 1 mM and Edaravone at concentrations 50 µM and 100 µM. Our data suggest that the hyperactive LRRK2 G2019S kinase activity leads to increase in KEAP1, which binds NRF2 and leads to its degradation, reduction in the antioxidant response, increased ROS, mitochondria dysfunction and cell death observed in the PD phenotype.


Subject(s)
Neural Stem Cells , Neurodegenerative Diseases , Parkinson Disease , Humans , Parkinson Disease/metabolism , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2/genetics , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2/metabolism , Antioxidants/pharmacology , Antioxidants/metabolism , Kelch-Like ECH-Associated Protein 1/metabolism , Neurodegenerative Diseases/metabolism , Reactive Oxygen Species/metabolism , Hydrogen Peroxide/metabolism , NF-E2-Related Factor 2/genetics , NF-E2-Related Factor 2/metabolism , Neural Stem Cells/metabolism
13.
Commun Biol ; 6(1): 431, 2023 04 19.
Article in English | MEDLINE | ID: mdl-37076591

ABSTRACT

Gaucher Disease (GD), the most common lysosomal disorder, arises from mutations in the GBA1 gene and is characterized by a wide spectrum of phenotypes, ranging from mild hematological and visceral involvement to severe neurological disease. Neuronopathic patients display dramatic neuronal loss and increased neuroinflammation, whose molecular basis are still unclear. Using a combination of Drosophila dGBA1b loss-of-function models and GD patient-derived iPSCs differentiated towards neuronal precursors and mature neurons we showed that different GD- tissues and neuronal cells display an impairment of growth mechanisms with an increased cell death and reduced proliferation. These phenotypes are coupled with the downregulation of several Hippo transcriptional targets, mainly involved in cells and tissue growth, and YAP exclusion from nuclei. Interestingly, Hippo knock-down in the GBA-KO flies rescues the proliferative defect, suggesting that targeting the Hippo pathway can be a promising therapeutic approach to neuronopathic GD.


Subject(s)
Gaucher Disease , Humans , Gaucher Disease/genetics , Gaucher Disease/metabolism , Gaucher Disease/therapy , Glucosylceramidase/genetics , Glucosylceramidase/metabolism , Hippo Signaling Pathway , Neurons/metabolism , Cell Proliferation
14.
Nat Commun ; 14(1): 1930, 2023 04 06.
Article in English | MEDLINE | ID: mdl-37024507

ABSTRACT

Mutations in GBA1, the gene encoding the lysosomal enzyme ß-glucocerebrosidase (GCase), which cause Gaucher's disease, are the most frequent genetic risk factor for Parkinson's disease (PD). Here, we employ global proteomic and single-cell genomic approaches in stable cell lines as well as induced pluripotent stem cell (iPSC)-derived neurons and midbrain organoids to dissect the mechanisms underlying GCase-related neurodegeneration. We demonstrate that GCase can be imported from the cytosol into the mitochondria via recognition of internal mitochondrial targeting sequence-like signals. In mitochondria, GCase promotes the maintenance of mitochondrial complex I (CI) integrity and function. Furthermore, GCase interacts with the mitochondrial quality control proteins HSP60 and LONP1. Disease-associated mutations impair CI stability and function and enhance the interaction with the mitochondrial quality control machinery. These findings reveal a mitochondrial role of GCase and suggest that defective CI activity and energy metabolism may drive the pathogenesis of GCase-linked neurodegeneration.


Subject(s)
Glucosylceramidase , Parkinson Disease , Humans , Glucosylceramidase/genetics , Glucosylceramidase/metabolism , Proteomics , Parkinson Disease/metabolism , Mitochondria/genetics , Mitochondria/metabolism , Energy Metabolism/genetics , Mutation , Lysosomes/metabolism , alpha-Synuclein/metabolism , Mitochondrial Proteins/metabolism , ATP-Dependent Proteases/metabolism
15.
Stem Cells ; 29(7): 1052-63, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21608081

ABSTRACT

Immune rejection and risk of tumor formation are perhaps the greatest hurdles in the field of stem cell transplantation. Here, we report the generation of several lines of induced pluripotent stem cells (iPSCs) from cynomolgus macaque (CM) skin fibroblasts carrying specific major histocompatibility complex (MHC) haplotypes. To develop a collection of MHC-matched iPSCs, we genotyped the MHC locus of 25 CMs by microsatellite polymerase chain reaction analysis. Using retroviral infection of dermal skin fibroblasts, we generated several CM-iPSC lines carrying different haplotypes. We characterized the immunological properties of CM-iPSCs and demonstrated that CM-iPSCs can be induced to differentiate in vitro along specific neuronal populations, such as midbrain dopaminergic (DA) neurons. Midbrain-like DA neurons generated from CM-iPSCs integrated into the striatum of a rodent model of Parkinson's disease and promoted behavioral recovery. Importantly, neither tumor formation nor inflammatory reactions were observed in the transplanted animals up to 6 months after transplantation. We believe that the generation and characterization of such histocompatible iPSCs will allow the preclinical validation of safety and efficacy of iPSCs for neurodegenerative diseases and several other human conditions in the field of regenerative medicine.


Subject(s)
Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/transplantation , Neurons/transplantation , Stem Cell Transplantation/methods , Animals , Cell Differentiation/physiology , Corpus Striatum/surgery , Female , Humans , Induced Pluripotent Stem Cells/immunology , Macaca fascicularis , Mice , Mice, Inbred NOD , Mice, SCID , Neurons/immunology , Parkinson Disease/immunology , Parkinson Disease/surgery , Rats , Rats, Sprague-Dawley
16.
NPJ Parkinsons Dis ; 8(1): 92, 2022 Jul 19.
Article in English | MEDLINE | ID: mdl-35853899

ABSTRACT

Leucine-rich repeat kinase 2 (LRRK2) is a kinase involved in different cellular functions, including autophagy, endolysosomal pathways, and immune function. Mutations in LRRK2 cause autosomal-dominant forms of Parkinson's disease (PD). Heterozygous mutations in GBA1, the gene encoding the lysosomal enzyme glucocerebrosidase (GCase), are the most common genetic risk factors for PD. Moreover, GCase function is altered in idiopathic PD and in other genetic forms of the disease. Recent work suggests that LRRK2 kinase activity can regulate GCase function. However, both a positive and a negative correlation have been described. To gain insights into the impact of LRRK2 on GCase, we performed a comprehensive analysis of GCase levels and activity in complementary LRRK2 models, including (i) LRRK2 G2019S knock in (GSKI) mice, (ii) peripheral blood mononuclear cell (PBMCs), plasma, and fibroblasts from PD patients carrying LRRK2 G2019S mutation, (iii) patient iPSCs-derived neurons; (iv) endogenous and overexpressed cell models. In some of these models we found a positive correlation between the activities of LRRK2 and GCase, which was further confirmed in cell lines with genetic and pharmacological manipulation of LRRK2 kinase activity. GCase protein level is reduced in GSKI brain tissues and in G2019S iPSCs-derived neurons, but increased in fibroblasts and PBMCs from patients, suggesting cell-type-specific effects. Overall, our study indicates that LRRK2 kinase activity affects both the levels and the catalytic activity of GCase in a cell-type-specific manner, with important implications in the context of therapeutic application of LRRK2 inhibitors in GBA1-linked and idiopathic PD.

17.
J Neurosci ; 30(48): 16091-101, 2010 Dec 01.
Article in English | MEDLINE | ID: mdl-21123556

ABSTRACT

In Parkinson's disease (PD), loss of striatal dopaminergic (DA) terminals and degeneration of DA neurons in the substantia nigra (SN) are associated with glial reactions. Such inflammatory processes are commonly considered an epiphenomenon of neuronal degeneration. However, there is increasing recognition of the role of neuroinflammation as an initiation factor of DA neuron degeneration. To investigate this issue, we established a new model of brain inflammation by injecting the Toll-like receptor 3 (TLR-3) agonist polyinosinic:polycytidylic acid [poly(I:C)] in the SN of adult rats. Poly(I:C) injection induced a sustained inflammatory reaction in the SN and in the dorsolateral striatum. Significant changes were detected in proteins relevant to synaptic transmission and axonal transport. In addition, cytoplasmic mislocalization of neuronal TAR DNA binding protein TDP-43 was observed. Poly(I:C) injection increased the susceptibility of midbrain DA neurons to a subsequent neurotoxic trigger (low-dose 6-hydroxydopamine). Systemic delivery of interleukin-1 receptor antagonist protected SN DA neurons exposed to combined poly(I:C) induced inflammatory and neurotoxic oxidative stress. These data indicate that viral-like neuroinflammation induces predegenerative changes in the DA system, which lowers the set point toward neuronal dysfunction and degeneration. New powerful neuroprotective therapies for PD might be considered by targeting critical inflammatory mechanisms, including cytokine-induced neurotoxicity.


Subject(s)
Corpus Striatum/pathology , Dopamine , Nerve Degeneration/pathology , Poly I-C/toxicity , Substantia Nigra/pathology , Toll-Like Receptor 3/agonists , Animals , Corpus Striatum/drug effects , Dopamine/physiology , Female , Injections, Intraventricular , Nerve Degeneration/chemically induced , Poly I-C/administration & dosage , Rats , Rats, Sprague-Dawley , Substantia Nigra/drug effects , Toll-Like Receptor 3/physiology
18.
Mol Cell Neurosci ; 45(3): 258-66, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20603216

ABSTRACT

The cardinal motor symptoms of Parkinson's disease (PD) are caused by the vulnerability to dysfunction and degeneration of ventral midbrain (VM) dopaminergic (DA) neurons. A major limitation for experimental studies of current ES/iPS cell differentiation protocols is the lack of VM DA neurons with a stable phenotype as defined by an expression marker code of FOXA2/TH/ß-tubulin. Here we demonstrate a combination of three modifications that were required to produce VM DA neurons. Firstly, early and specific exposure to 10(-)(8)M (low dose) retinoic acid improved the regional identity of neural progenitor cells derived from human ES cells, PD or healthy subject-specific iPS cells. Secondly, a high activity form of human sonic hedgehog established a sizeable FOXA2(+) neural progenitor cell population in vitro. Thirdly, early exposure to FGF8a, rather than Fgf8b, and WNT1 was required for robust differentiation of the FOXA2(+) floor plate-like human neural progenitor cells into FOXA2(+) DA neurons. FOXA2(+) DA neurons were also generated when this protocol was adapted to feeder-free conditions. In summary, this new human ES and iPS cell differentiation protocol using FGF8a, WNT1, low dose retinoic acid and a high activity form of SHH can generate human VM DA neurons that are required for relevant new bioassays, drug discovery and cell based therapies for PD.


Subject(s)
Cell Differentiation/drug effects , Dopamine/metabolism , Embryonic Stem Cells/cytology , Fibroblast Growth Factor 8/pharmacology , Hedgehog Proteins/metabolism , Neurons/cytology , Pluripotent Stem Cells/cytology , Tretinoin/pharmacology , Animals , Cell Culture Techniques , Cell Differentiation/physiology , Cells, Cultured , Embryonic Stem Cells/drug effects , Embryonic Stem Cells/physiology , Hedgehog Proteins/genetics , Hepatocyte Nuclear Factor 3-beta/metabolism , Humans , Mesencephalon/cytology , Mice , Neurons/drug effects , Neurons/metabolism , Parkinson Disease , Pluripotent Stem Cells/drug effects , Pluripotent Stem Cells/physiology , Wnt1 Protein/pharmacology
19.
Trends Neurosci ; 44(10): 771-780, 2021 10.
Article in English | MEDLINE | ID: mdl-34284880

ABSTRACT

The antagonistic pleiotropy (AP) theory posits that adaptive evolutionary changes, which facilitate reproduction and individual fitness early in life, can enhance detrimental aging-related processes. Several genes associated with human brain diseases play a protective role in infection, suggesting the relevance of AP in the context of brain aging and neurodegeneration. Relatedly, genetic variants that confer immune protection against pathogens may lead to uncontrolled brain inflammation later in life. Here, we propose a conceptual framework suggesting that the pleiotropic roles of genes in infections and host-pathogen interactions should be considered when studying neurological illnesses. We reinterpret recent findings regarding the impact of neurological disease-associated genetic traits on infections and chronic inflammatory diseases. Identifying the AP pathways shared among these seemingly unrelated conditions might provide further insights into the detrimental role of the immune system in brain disease as well as the mechanisms involved in chronic infections.


Subject(s)
Neurodegenerative Diseases , Aging , Biological Evolution , Brain , Humans , Neurodegenerative Diseases/genetics , Phenotype
20.
Mech Ageing Dev ; 197: 111499, 2021 07.
Article in English | MEDLINE | ID: mdl-33989633

ABSTRACT

The decline of nicotinamide adenine dinucleotide (NAD+) levels is a hallmark of aging in multiple organisms and tissues, including the human brain. Hence, agents that increase intracellular NAD+ could have beneficial effects in aging and age-related neurodegenerative diseases. Disturbances in NAD+ metabolism have also been observed in Parkinson's disease (PD), supporting a link between neuronal bioenergetics failure and disease pathogenesis. Here, we review emerging findings revealing key roles for NAD+ and related metabolites in experimental models of dopaminergic neurodegeneration and in PD patients. We discuss how increased NAD+ levels might ameliorate disease phenotypes by restoring neuronal mitochondrial energy metabolism, promoting cellular proteostasis, and modulating the immune system. Finally, we describe ongoing clinical trials targeting NAD+ in PD and highlight the need for further investigations to better delineate the association between NAD+, brain aging and disease, and optimal strategies for efficiently and safely raising NAD+ levels. A more comprehensive understanding of the basic mechanisms linking NAD+, energy metabolism, and PD, and of the impact of life-long NAD+ targeting strategies, are critical to inform future clinical applications.


Subject(s)
Aging/metabolism , Energy Metabolism , NAD/metabolism , Parkinson Disease/metabolism , Parkinson Disease/therapy , Animals , Clinical Trials as Topic , Humans
SELECTION OF CITATIONS
SEARCH DETAIL