scispace - formally typeset
Search or ask a question

Showing papers by "Valina L. Dawson published in 2022"


Journal ArticleDOI
TL;DR: The results suggest that STING inhibition may be therapeutic in idiopathic PD and possibly other human α-synucleinopathies, and it is shown that αSyn aggregates can increase STING expression and augment canonical STING activation, suggesting a possible generalized propensity for exaggerated antiviral responses in neurological states with STING elevation.
Abstract: Significance It is increasingly recognized that chronic neuroinflammation is causally relevant to neurodegeneration. In Parkinson’s disease (PD), α-synuclein pathology activates inflammatory signaling that disturbs parenchymal homeostasis and disrupts neuron-glia interactions. Herein, we report that the innate immune cyclic GMP-AMP synthase (cGAS)/stimulator of interferon genes (STING) DNA-sensing pathway is activated in a mouse model of α-synucleinopathy and parkinsonism, leading to type-I interferon activation that precedes the onset of neurodegeneration. Remarkably, STING-deficient mice were protected from dopaminergic neuron loss in this model. We also show that αSyn aggregates can increase STING expression and augment canonical STING activation, suggesting a possible generalized propensity for exaggerated antiviral responses in neurological states with STING elevation. Our results suggest that STING inhibition may be therapeutic in idiopathic PD and possibly other human α-synucleinopathies.

36 citations


Journal ArticleDOI
25 May 2022-Neuron
TL;DR: In this paper , Parkin activation was found to contribute to the assembly of an active NLRP3 inflammasome complex via mitochondrial-derived reactive oxygen species (mitoROS) generation through the accumulation of another parkin ubiquitination substrate, ZNF746/PARIS.

34 citations


Journal ArticleDOI
01 May 2022-Cell
TL;DR: It is shown that neurodegeneration induced by pathologic α-synuclein (α-syn) occurs via PAAN/MIF nuclease activity and could have broad relevance in human pathologies where parthanatos plays a role in the development of cell death inhibitors targeting the druggable PAAN/.

15 citations


Journal ArticleDOI
TL;DR: It is shown that irisin prevents the accumulation of pathologic α-syn and neuronal cell death by enhancing endolysosomal degradation of pathological α- synuclein in PD.
Abstract: Significance Physical exercise is thought to have beneficial effects on the symptoms of Parkinson’s disease (PD). Irisin is an exercise-induced myokine released into the circulation. We therefore tested whether irisin itself could have a beneficial effect on pathologic α-synuclein (α-syn) accumulation and concomitant neurodegeneration in PD. Here, we show that irisin prevents the accumulation of pathologic α-syn and neuronal cell death by enhancing endolysosomal degradation of pathologic α-syn. Furthermore, elevation of blood irisin levels in mice prevented neurodegeneration and physiological deficits induced by injection α-syn preformed fibrils. These findings would seem to have translational promise as a disease-modifying therapy for treating PD and other neurodegenerative diseases involving pathologic α-syn.

10 citations


Journal ArticleDOI
TL;DR: Sveinbjorndottir et al. as discussed by the authors showed that misfolded α-syn activates microglia, which release interleukin 6 (IL-6).

9 citations


Journal ArticleDOI
TL;DR: CYLD is identified as a negative regulator of dopamine neuron survival, and inhibition of CYLD may potentially be beneficial in PD by lowering PARIS levels and promoting mitochondrial biogenesis.
Abstract: Mutations in PINK1 and parkin highlight the mitochondrial axis of Parkinson's disease (PD) pathogenesis. PINK1/parkin regulation of the transcriptional repressor PARIS bears direct relevance to dopamine neuron survival through augmentation of PGC-1α-dependent mitochondrial biogenesis. Notably, knockout of PARIS attenuates dopaminergic neurodegeneration in mouse models, indicating that interventions that prevent dopaminergic accumulation of PARIS could have therapeutic potential in PD. To this end, we have identified the deubiquitinase cylindromatosis (CYLD) to be a regulator of PARIS protein stability and proteasomal degradation via the PINK1/parkin pathway. Knockdown of CYLD in multiple models of PINK1 or parkin inactivation attenuates PARIS accumulation by modulating its ubiquitination levels and relieving its repressive effect on PGC-1α to promote mitochondrial biogenesis. Together, our studies identify CYLD as a negative regulator of dopamine neuron survival, and inhibition of CYLD may potentially be beneficial in PD by lowering PARIS levels and promoting mitochondrial biogenesis.

8 citations


Journal ArticleDOI
TL;DR: It is shown that loss of poly(ADP-ribose) (PAR) suppresses neurodegeneration in c9ALS/FTD fly models and neurons differentiated from patient-derived induced pluripotent stem cells.
Abstract: Arginine-rich dipeptide repeat proteins (R-DPRs), abnormal translational products of a GGGGCC hexanucleotide repeat expansion in C9ORF72, play a critical role in C9ORF72-related amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD), the most common genetic form of the disorders (c9ALS/FTD). R-DPRs form liquid condensates in vitro, induce stress granule formation in cultured cells, aggregate, and sometimes coaggregate with TDP-43 in postmortem tissue from patients with c9ALS/FTD. However, how these processes are regulated is unclear. Here, we show that loss of poly(ADP-ribose) (PAR) suppresses neurodegeneration in c9ALS/FTD fly models and neurons differentiated from patient-derived induced pluripotent stem cells. Mechanistically, PAR induces R-DPR condensation and promotes R-DPR–induced stress granule formation and TDP-43 aggregation. Moreover, PAR associates with insoluble R-DPR and TDP-43 in postmortem tissue from patients. These findings identified PAR as a promoter of R-DPR toxicity and thus a potential target for treating c9ALS/FTD. Description Poly(ADP-ribose) promotes the condensation and toxicity of C9ORF72 arginine-rich dipeptide repeat proteins and contributes to neurodegeneration. Bringing the target up to PAR The most common genetic form of amyotrophic lateral sclerosis/frontotemporal dementia (ALS/LTD) is caused by repeat expansion of the hexanucleotide G4C2 in the C9ORF72 gene, the subsequent production of arginine-rich dipeptide repeat proteins (R-DPRs) ultimately resulting in neurodegeneration. Here, Gao et al. investigated how R-DPR formation could lead to neuronal loss and showed that the polymer poly(ADP)-ribose (PAR) interacts with R-DPR and promotes stress granule formation and TDP-43 aggregation in fly models. Similar effects were found in samples from patients with ALS/FTD, suggesting that targeting PAR could reduce the deleterious effects of R-DPR in C9ORF72-mediated ALS/FTD.

6 citations


Journal ArticleDOI
TL;DR: It is shown that Parkin-/-/PolgAD257A/D257A mice, a previously reported PD mouse model, fails to reproduce a Parkinsonian phenotype and is reported that parkin loss does not synergize with mitochondrial dysfunction in mouse models of mitochondrial deficits.
Abstract: Parkinson's disease (PD) is characterized by the progressive loss of dopaminergic neurons in the substantia nigra pars compacta (SNpc). In this study, we generated a transgenic model by crossing germline Parkin–/– mice with PolgAD257A mice, an established model of premature aging and mitochondrial stress. We hypothesized that loss of Parkin–/– in PolgAD257A/D257A mice would exacerbate mitochondrial dysfunction, leading to loss of dopamine neurons and nigral-striatal specific neurobehavioral motor dysfunction. We found that aged Parkin–/–/PolgAD257A/D257A male and female mice exhibited severe behavioral deficits, nonspecific to the nigral-striatal pathway, with neither dopaminergic neurodegeneration nor reductions in striatal dopamine. We saw no difference in expression levels of nuclear-encoded subunits of mitochondrial markers and mitochondrial Complex I and IV activities, although we did observe substantial reductions in mitochondrial-encoded COX41I, indicating mitochondrial dysfunction as a result of PolgAD257A/D257A mtDNA mutations. Expression levels of mitophagy markers LC3I/LC3II remained unchanged between cohorts, suggesting no overt mitophagy defects. Expression levels of the parkin substrates, VDAC, NLRP3, and AIMP2 remained unchanged, suggesting no parkin dysfunction. In summary, we were unable to observe dopaminergic neurodegeneration with corresponding nigral-striatal neurobehavioral deficits, nor Parkin or mitochondrial dysfunction in Parkin–/–/PolgAD257A/D257A mice. These findings support a lack of synergism of Parkin loss on mitochondrial dysfunction in mouse models of mitochondrial deficits. SIGNIFICANCE STATEMENT Producing a mouse model of Parkinson's disease (PD) that is etiologically relevant, recapitulates clinical hallmarks, and exhibits reproducible results is crucial to understanding the underlying pathology and in developing disease-modifying therapies. Here, we show that Parkin–/–/PolgAD257A/D257A mice, a previously reported PD mouse model, fails to reproduce a Parkinsonian phenotype. We show that these mice do not display dopaminergic neurodegeneration nor nigral-striatal-dependent motor deficits. Furthermore, we report that Parkin loss does not synergize with mitochondrial dysfunction. Our results demonstrate that Parkin–/–/PolgAD257A/D257A mice are not a reliable model for PD and adds to a growing body of work demonstrating that Parkin loss does not synergize with mitochondrial dysfunction in mouse models of mitochondrial deficits.

4 citations



Journal ArticleDOI
TL;DR: This research presents a novel and scalable approach to regenerative medicine that combines cell reprograming, which has the potential to improve the quality of life of patients and also contributes to their prognosis.
Abstract: 1Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA 2Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA 3Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA 4Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA 5Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA

2 citations


Journal ArticleDOI
TL;DR: In this article , the AAA + ATPase Thorase directly binds mTOR, thereby orchestrating the disassembly and inactivation of mTORC1. Thorase disrupts the association of m TOR to Raptor at the mitochondria-lysosome interface and this action is sensitive to amino acids.
Abstract: The mechanistic target of rapamycin (mTOR) signals through the mTOR complex 1 (mTORC1) and the mTOR complex 2 to maintain cellular and organismal homeostasis. Failure to finely tune mTOR activity results in metabolic dysregulation and disease. While there is substantial understanding of the molecular events leading mTORC1 activation at the lysosome, remarkably little is known about what terminates mTORC1 signaling. Here, we show that the AAA + ATPase Thorase directly binds mTOR, thereby orchestrating the disassembly and inactivation of mTORC1. Thorase disrupts the association of mTOR to Raptor at the mitochondria-lysosome interface and this action is sensitive to amino acids. Lack of Thorase causes accumulation of mTOR-Raptor complexes and altered mTORC1 disassembly/re-assembly dynamics upon changes in amino acid availability. The resulting excessive mTORC1 can be counteracted with rapamycin in vitro and in vivo. Collectively, we reveal Thorase as a key component of the mTOR pathway that disassembles and thus inhibits mTORC1.

Posted ContentDOI
06 Jun 2022-bioRxiv
TL;DR: The global database of PAR-binding proteins that is established will be a valuable tool for further in-depth analysis of the role of PARylation in a wide range of biological contexts.
Abstract: Poly(ADP-ribose) (PAR) plays a crucial role in intracellular signaling and scaffolding through covalent modification or non-covalent binding to target proteins. The non- covalent binding PARylome has not been extensively characterized. Here we performed a PAR-binding screen using a human protein microarray that covers most of the human proteome to characterize the non-covalent binding PARylome. A total of 356 PAR- binding proteins were identified. The PAR-binding PARylome suggests that PAR- binding regulates a variety of biological processes beyond well-characterized DNA damage signaling and DNA repair. Proteins that may be reprogrammed by PAR-binding include signaling molecules, transcription factors, nucleic acid binding proteins, calcium binding proteins, ligases, oxidoreductases, enzymes, transferases, hydrolases, and receptors. The global database of PAR-binding proteins that we established will be a valuable tool for further in-depth analysis of the role of PARylation in a wide range of biological contexts.

Journal ArticleDOI
TL;DR: This article identified a high-affinity cocaine binding site associated with BASP1 that is involved in mediating the drug's psychotropic actions, suggesting the existence of a high affinity receptor(s) for the drug.
Abstract: Significance Cocaine is a monoamine transport inhibitor. Current models attributing pharmacologic actions of cocaine to inhibiting the activity of the amine transporters alone failed to translate to the clinic. Cocaine inhibition of the dopamine, serotonin, and norepinephrine transporters is relatively weak, suggesting that blockade of the amine transporters alone cannot account for the actions of cocaine, especially at low doses. There is evidence for significantly more potent actions of cocaine, suggesting the existence of a high-affinity receptor(s) for the drug. Identifying and characterizing such receptors will deepen our understanding of cocaine pharmacologic actions and pave the way for therapeutic development. Here we identify a high-affinity cocaine binding site associated with BASP1 that is involved in mediating the drug’s psychotropic actions.

Posted ContentDOI
24 Nov 2022-bioRxiv
TL;DR: In this paper , the PAR-binding motif in the huntingtin protein was measured by fluorescence polarization and visualized by atomic force microscopy, and the results provided insight into a very early molecular mechanism of Alzheimer's and Parkinson's diseases.
Abstract: Huntington’s disease (HD) is an autosomal dominant genetic neurodegenerative disease caused by a CAG expansion in the Huntingtin (HTT) gene, translating to an expanded polyglutamine tract in the huntingtin (HTT) protein. Age at disease onset is correlated to CAG repeat length, but varies by decades between individuals with identical repeat lengths. Genome-wide association studies link HD modification to DNA repair and mitochondrial health pathways. Recent clinical studies show elevated DNA damage in HD, even at the premanifest stage of disease. One of the major DNA repair nodes influencing neurodegenerative disease is the PARP pathway. Accumulation of poly ADP-ribose (PAR), produced by PARP1 and PARP2, has been implicated in the pathology of Alzheimer’s and Parkinson’s diseases, as well as autosomal recessive cerebellar ataxia. We report that HD mutation carriers have lower cerebrospinal fluid PAR levels than healthy controls, starting at the premanifest stage. Patient-derived fibroblasts have reduced PARP1/2 activity and elevated DNA damage, while elevated PAR levels are only revealed upon inhibition of PAR degradation. These phenotypes are rescued by moderate huntingtin level reduction via the huntingtin-lowering splice modulator drug, LMI070 (Branaplam). As a direct mechanism, we have defined a PAR-binding motif in huntingtin, detected huntingtin complexed with PARylated proteins in human cells during stress, and localized huntingtin to mitotic chromosomes upon inhibition of PAR degradation. Direct huntingtin PAR binding was measured by fluorescence polarization and visualized by atomic force microscopy. These results provide insight into a very early molecular mechanism of HD, suggesting possible targets in HD to design early preventive therapies.

Posted ContentDOI
06 Oct 2022-bioRxiv
TL;DR: It is reported that pathologic α-syn binds to tuberous sclerosis protein (TSC) 2 and destabilizes the TSC1-TSC2 complex leading to activation of the mechanistic target of rapamycin (mTOR) complex 1 ( mTORC1) and enhanced mRNA translation.
Abstract: Pathologic α-syn destabilizes the TSC 1 and 2 complex leading to mTORC1 activation, enhanced protein translation and neurodegeneration in PD. Abstract: Pathological α-synuclein (α-syn) plays an important role in the pathogenesis of α-synucleinopathies such as Parkinson’s disease (PD). Disruption of protein homeostasis is thought be central to PD pathogenesis, however the molecular mechanism of this deregulation is poorly understood. Here we report that pathologic α-syn binds to tuberous sclerosis protein (TSC) 2 and destabilizes the TSC1-TSC2 complex leading to activation of the mechanistic target of rapamycin (mTOR) complex 1 (mTORC1) and enhanced mRNA translation. Dopamine neuron loss, behavioral deficits and aberrant biochemical signaling in the α-syn preformed fibril (PFF) and Drosophila α-syn transgenic models of pathologic α-syn induced degeneration were attenuated by genetic and pharmacologic inhibition of mTOR and protein translation. Our findings establish a potential molecular mechanism by which pathologic α-syn activates mTORC1 leading to enhanced protein translation and concomitant neurodegeneration in PD.