Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Cells ; 13(3)2024 Jan 29.
Article in English | MEDLINE | ID: mdl-38334646

ABSTRACT

Protein aggregation is a predominant feature of many neurodegenerative diseases, including synucleinopathies, which are characterized by cellular inclusions containing α-Synuclein (αSyn) phosphorylated at serine 129 (pSer129). In the present study, we characterized the development of αSyn pre-formed fibril (PFF)-induced pSer129-αSyn pathology in F28tg mice overexpressing human wild-type αSyn, as well as in ex vivo organotypic cultures and in vitro primary cultures from the same mouse model. Concurrently, we collected cerebrospinal fluid (CSF) from mice and conditioned media from ex vivo and in vitro cultures and quantified the levels of neurofilament light chain (NFL), a biomarker of neurodegeneration. We found that the intra-striatal injection of PFFs induces the progressive spread of pSer129-αSyn pathology and microglial activation in vivo, as well as modest increases in NFL levels in the CSF. Similarly, PFF-induced αSyn pathology occurs progressively in ex vivo organotypic slice cultures and is accompanied by significant increases in NFL release into the media. Using in vitro primary hippocampal cultures, we further confirmed that pSer129-αSyn pathology and NFL release occur in a manner that correlates with the fibril dose and the level of the αSyn protein. Overall, we demonstrate that αSyn pathology is associated with NFL release across preclinical models of seeded αSyn aggregation and that the pharmacological inhibition of αSyn aggregation in vitro also significantly reduces NFL release.


Subject(s)
Neurodegenerative Diseases , Synucleinopathies , Animals , Humans , Mice , alpha-Synuclein/metabolism , Intermediate Filaments/metabolism , Neurodegenerative Diseases/pathology , Protein Aggregates/physiology
2.
Cell Death Dis ; 15(1): 52, 2024 01 15.
Article in English | MEDLINE | ID: mdl-38225227

ABSTRACT

Ubiquitination of mitochondrial proteins plays an important role in the cellular regulation of mitophagy. The E3 ubiquitin ligase parkin (encoded by PARK2) and the ubiquitin-specific protease 30 (USP30) have both been reported to regulate the ubiquitination of outer mitochondrial proteins and thereby mitophagy. Loss of E3 ligase activity is thought to be pathogenic in both sporadic and inherited Parkinson's disease (PD), with loss-of-function mutations in PARK2 being the most frequent cause of autosomal recessive PD. The aim of the present study was to evaluate whether mitophagy induced by USP30 inhibition provides a functional rescue in isogenic human induced pluripotent stem cell-derived dopaminergic neurons with and without PARK2 knockout (KO). Our data show that healthy neurons responded to CCCP-induced mitochondrial damage by clearing the impaired mitochondria and that this process was accelerated by USP30 inhibition. Parkin-deficient neurons showed an impaired mitophagic response to the CCCP challenge, although mitochondrial ubiquitination was enhanced. USP30 inhibition promoted mitophagy in PARK2 KO neurons, independently of whether left in basal conditions or treated with CCCP. In PARK2 KO, as in control neurons, USP30 inhibition balanced oxidative stress levels by reducing excessive production of reactive oxygen species. Interestingly, non-dopaminergic neurons were the main driver of the beneficial effects of USP30 inhibition. Our findings demonstrate that USP30 inhibition is a promising approach to boost mitophagy and improve cellular health, also in parkin-deficient cells, and support the potential relevance of USP30 inhibitors as a novel therapeutic approach in diseases with a need to combat neuronal stress mediated by impaired mitochondria.


Subject(s)
Induced Pluripotent Stem Cells , Oxidative Stress , Parkinsonian Disorders , Ubiquitin-Protein Ligases , Humans , Carbonyl Cyanide m-Chlorophenyl Hydrazone/adverse effects , Dopaminergic Neurons/metabolism , Induced Pluripotent Stem Cells/metabolism , Mitochondrial Proteins/metabolism , Mitophagy , Parkinsonian Disorders/pathology , Ubiquitin-Protein Ligases/genetics
3.
Mol Neurodegener ; 16(1): 54, 2021 08 11.
Article in English | MEDLINE | ID: mdl-34380535

ABSTRACT

BACKGROUND: Proteopathic brain lesions are a hallmark of many age-related neurodegenerative diseases including synucleinopathies and develop at least a decade before the onset of clinical symptoms. Thus, understanding of the initiation and propagation of such lesions is key for developing therapeutics to delay or halt disease progression. METHODS: Alpha-synuclein (αS) inclusions were induced in long-term murine and human slice cultures by seeded aggregation. An αS seed-recognizing human antibody was tested for blocking seeding and/or spreading of the αS lesions. Release of neurofilament light chain (NfL) into the culture medium was assessed. RESULTS: To study initial stages of α-synucleinopathies, we induced αS inclusions in murine hippocampal slice cultures by seeded aggregation. Induction of αS inclusions in neurons was apparent as early as 1week post-seeding, followed by the occurrence of microglial inclusions in vicinity of the neuronal lesions at 2-3 weeks. The amount of αS inclusions was dependent on the type of αS seed and on the culture's genetic background (wildtype vs A53T-αS genotype). Formation of αS inclusions could be monitored by neurofilament light chain protein release into the culture medium, a fluid biomarker of neurodegeneration commonly used in clinical settings. Local microinjection of αS seeds resulted in spreading of αS inclusions to neuronally connected hippocampal subregions, and seeding and spreading could be inhibited by an αS seed-recognizing human antibody. We then applied parameters of the murine cultures to surgical resection-derived adult human long-term neocortical slice cultures from 22 to 61-year-old donors. Similarly, in these human slice cultures, proof-of-principle induction of αS lesions was achieved at 1week post-seeding in combination with viral A53T-αS expressions. CONCLUSION: The successful translation of these brain cultures from mouse to human with the first reported induction of human αS lesions in a true adult human brain environment underlines the potential of this model to study proteopathic lesions in intact mouse and now even aged human brain environments.


Subject(s)
Microglia/pathology , Neurofilament Proteins/metabolism , Neurons/pathology , Organ Culture Techniques/methods , Synucleinopathies , Animals , Humans , Inclusion Bodies/pathology , Mice , Microglia/metabolism , Neurons/metabolism , alpha-Synuclein/toxicity
4.
Cell ; 163(2): 324-39, 2015 Oct 08.
Article in English | MEDLINE | ID: mdl-26451483

ABSTRACT

Neurodegenerative diseases have been linked to inflammation, but whether altered immunomodulation plays a causative role in neurodegeneration is not clear. We show that lack of cytokine interferon-ß (IFN-ß) signaling causes spontaneous neurodegeneration in the absence of neurodegenerative disease-causing mutant proteins. Mice lacking Ifnb function exhibited motor and cognitive learning impairments with accompanying α-synuclein-containing Lewy bodies in the brain, as well as a reduction in dopaminergic neurons and defective dopamine signaling in the nigrostriatal region. Lack of IFN-ß signaling caused defects in neuronal autophagy prior to α-synucleinopathy, which was associated with accumulation of senescent mitochondria. Recombinant IFN-ß promoted neurite growth and branching, autophagy flux, and α-synuclein degradation in neurons. In addition, lentiviral IFN-ß overexpression prevented dopaminergic neuron loss in a familial Parkinson's disease model. These results indicate a protective role for IFN-ß in neuronal homeostasis and validate Ifnb mutant mice as a model for sporadic Lewy body and Parkinson's disease dementia.


Subject(s)
Interferon-beta/metabolism , Neurons/metabolism , Receptor, Interferon alpha-beta/metabolism , Animals , Autophagy , Disease Models, Animal , Genetic Therapy , Interferon-beta/genetics , Interferon-beta/therapeutic use , Lewy Body Disease/metabolism , Lewy Body Disease/pathology , Mice , Mice, Inbred C57BL , Parkinson Disease/genetics , Parkinson Disease/metabolism , Parkinson Disease/pathology , Parkinson Disease/therapy , Receptor, Interferon alpha-beta/genetics , Signal Transduction , Transcriptome , alpha-Synuclein/metabolism
5.
J Neurosci ; 33(35): 14231-45, 2013 Aug 28.
Article in English | MEDLINE | ID: mdl-23986257

ABSTRACT

Glioblastoma multiforme (GBM) is the most aggressive form of brain tumor. In general, tumor growth requires disruption of the tissue microenvironment, yet how this affects glioma progression is unknown. We studied program death-ligand (PD-L)1 in neurons and gliomas in tumors from GBM patients and associated the findings with clinical outcome. Remarkably, we found that upregulation of PD-L1 by neurons in tumor-adjacent brain tissue (TABT) associated positively with GBM patient survival, whereas lack of neuronal PD-L1 expression was associated with high PD-L1 in tumors and unfavorable prognosis. To understand the molecular mechanism of PD-L1 signaling in neurons, we investigated PD-L1 function in cerebellar and cortical neurons and its impact on gliomas. We discovered that neuronal PD-L1-induced caspase-dependent apoptosis of glioma cells. Because interferon (IFN)-ß induces PD-L1 expression, we studied the functional consequences of neuronal Ifnb gene deletion on PD-L1 signaling and function. Ifnb-/- neurons lacked PD-L1 and were defective in inducing glioma cell death; this effect was reversed on PD-L1 gene transfection. Ifnb-/- mice with intracerebral isografts survived poorly. Similar to the observations in GBM patients, better survival in wild-type mice was associated with high neuronal PD-L1 in TABT and downregulation of PD-L1 in tumors, which was defective in Ifnb-/- mice. Our data indicated that neuronal PD-L1 signaling in brain cells was important for GBM patient survival. Reciprocal PD-L1 regulation in TABT and tumor tissue could be a prognostic biomarker for GBM. Understanding the complex interactions between tumor and adjacent stromal tissue is important in designing targeted GBM therapies.


Subject(s)
B7-H1 Antigen/genetics , Biomarkers, Tumor/genetics , Brain Neoplasms/metabolism , Glioblastoma/metabolism , Neurons/metabolism , Adult , Aged , Animals , Apoptosis , B7-H1 Antigen/metabolism , Biomarkers, Tumor/metabolism , Brain Neoplasms/diagnosis , Cerebellum/pathology , Cerebral Cortex/pathology , Female , Gene Deletion , Gene Expression Regulation, Neoplastic , Glioblastoma/diagnosis , Humans , Interferon-beta/genetics , Interferon-beta/metabolism , Male , Mice , Middle Aged , Prognosis
6.
PLoS One ; 8(6): e65371, 2013.
Article in English | MEDLINE | ID: mdl-23785422

ABSTRACT

Alterations in function of the neurotrophin BDNF are associated with neurodegeneration, cognitive decline, and psychiatric disorders. BDNF promotes axonal outgrowth and branching, regulates dendritic tree morphology and is important for axonal regeneration after injury, responses that largely result from activation of its tyrosine kinase receptor TrkB. Although intracellular neurotrophin (NT) signaling presumably reflects the combined action of kinases and phosphatases, little is known about the contributions of the latter to TrkB regulation. The issue is complicated by the fact that phosphatases belong to multiple independently evolved families, which are rarely studied together. We undertook a loss-of-function RNA-interference-based screen of virtually all known (254) human phosphatases to understand their function in BDNF/TrkB-mediated neurite outgrowth in differentiated SH-SY5Y cells. This approach identified phosphatases from diverse families, which either positively or negatively modulate BDNF-TrkB-mediated neurite outgrowth, and most of which have little or no previously established function related to NT signaling. "Classical" protein tyrosine phosphatases (PTPs) accounted for 13% of the candidate regulatory phosphatases. The top classical PTP identified as a negative regulator of BDNF-TrkB-mediated neurite outgrowth was PTPN12 (also called PTP-PEST). Validation and follow-up studies showed that endogenous PTPN12 antagonizes tyrosine phosphorylation of TrkB itself, and the downstream activation of ERK1/2. We also found PTPN12 to negatively regulate phosphorylation of p130cas and FAK, proteins with previously described functions related to cell motility and growth cone behavior. Our data provide the first comprehensive survey of phosphatase function in NT signaling and neurite outgrowth. They reveal the complexity of phosphatase control, with several evolutionarily unrelated phosphatase families cooperating to affect this biological response, and hence the relevance of considering all phosphatase families when mining for potentially druggable targets.


Subject(s)
Neurites/physiology , Phosphoric Monoester Hydrolases/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 12/metabolism , Receptor, trkB/metabolism , Brain-Derived Neurotrophic Factor/pharmacology , Cell Differentiation/drug effects , Cell Line , Crk-Associated Substrate Protein/metabolism , Drug Evaluation, Preclinical/methods , Focal Adhesion Kinase 1/metabolism , Gene Knockdown Techniques , Humans , Neurites/drug effects , Neurons/cytology , Neurons/drug effects , Neurons/metabolism , Phenotype , Phosphoric Monoester Hydrolases/genetics , Phosphorylation , Protein Tyrosine Phosphatase, Non-Receptor Type 12/genetics , RNA Interference , Reproducibility of Results , Signal Transduction , Tretinoin/pharmacology
7.
Autophagy ; 9(3): 287-302, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23221969

ABSTRACT

IFNB1/interferon (IFN)-ß belongs to the type I IFNs and exerts potent antiproliferative, proapoptotic, antiangiogenic and immunemodulatory functions. Despite the beneficial effects of IFNB1 in experimental breast cancers, clinical translation has been disappointing, possibly due to induction of survival pathways leading to treatment resistance. Defects in autophagy, a conserved cellular degradation pathway, are implicated in numerous cancer diseases. Autophagy is induced in response to cancer therapies and can contribute to treatment resistance. While the type II IFN, IFNG, which in many aspects differs significantly from type I IFNs, can induce autophagy, no such function for any type I IFN has been reported. We show here that IFNB1 induces autophagy in MCF-7, MDAMB231 and SKBR3 breast cancer cells by measuring the turnover of two autophagic markers, MAP1LC3B/LC3 and SQSTM1/p62. The induction of autophagy in MCF-7 cells occurred upstream of the negative regulator of autophagy MTORC1, and autophagosome formation was dependent on the known core autophagy molecule ATG7 and the IFNB1 signaling molecule STAT1. Using siRNA-mediated silencing of several core autophagy molecules and STAT1, we provide evidence that IFNB1 mediates its antiproliferative effects independent of autophagy, while the proapoptotic function of IFNB1 was strongly enhanced in the absence of autophagy. This suggests that autophagy induced by IFNB1 promoted survival, which might contribute to tumor resistance against IFNB1 treatment. It may therefore be clinically relevant to reconcile a role for IFNB1 in the treatment of breast cancer with concomitant inhibition of autophagy.


Subject(s)
Apoptosis , Autophagy , Breast Neoplasms/metabolism , Gene Expression Regulation, Neoplastic , Interferon-beta/pharmacology , Animals , Cell Cycle , Cell Line, Tumor , Cell Survival , Female , Fibroblasts/metabolism , Humans , MCF-7 Cells , Mice , STAT1 Transcription Factor/metabolism , Signal Transduction
8.
Eur J Cell Biol ; 88(8): 433-43, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19446362

ABSTRACT

Metallothionein (MT) is a metal-binding protein capable of preventing oxidative stress and apoptotic cell death in the central nervous system of mammals, and hence is of putative therapeutic value in the treatment of neurodegenerative disorders. Recently, we demonstrated that a peptide modeled after the beta-domain of MT, EmtinB, induced neurite outgrowth and increased neuronal survival through binding to receptors of the low-density lipoprotein receptor family (LDLR). The present study identified two MT alpha-domain-derived peptide sequences termed EmtinAn and EmtinAc, each consisting of 14 amino acids, as potent stimulators of neuronal differentiation and survival of primary neurons. In addition, we show that a peptide derived from the N-terminus of the MT beta-domain, EmtinBn, promotes neuronal survival. The neuritogenic and survival promoting effects of EmtinAc, similar to MT and EmtinB but not EmtinAn, were dependent on the functional integrity of LDLR. Moreover, EmtinAn and EmtinAc induced activation of extracellular signal-regulated kinase (ERK) and protein kinase B (PKB/Akt). We suggest that multiple functional sites of MT serve to cross-link MT receptor(s), thereby transducing signals leading to an increase in neurite outgrowth and survival.


Subject(s)
Cerebellum/cytology , Metallothionein/chemistry , Neurites , Peptides , Protein Structure, Tertiary , Amino Acid Sequence , Animals , Apoptosis/drug effects , Cell Survival/drug effects , Cerebellum/drug effects , Cerebellum/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Molecular Sequence Data , Neurites/drug effects , Neurites/physiology , Peptides/chemical synthesis , Peptides/pharmacology , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Rats , Rats, Wistar , Receptors, Lipoprotein/metabolism
9.
J Neurochem ; 104(1): 21-37, 2008 Jan.
Article in English | MEDLINE | ID: mdl-17986228

ABSTRACT

Accumulating evidence suggests that metallothionein (MT)-I and -II promote neuronal survival and regeneration in vivo. The present study investigated the molecular mechanisms underlying the differentiation and survival-promoting effects of MT and a peptide modeled after MT, EmtinB. Both MT and EmtinB directly stimulated neurite outgrowth and promoted survival in vitro using primary cultures of cerebellar granule neurons. In addition, expression and surface localization of megalin, a known MT receptor, and the related lipoprotein receptor-related protein-1 (LRP) are demonstrated in cerebellar granule neurons. By means of surface plasmon resonance MT and EmtinB were found to bind to both megalin and LRP. The bindings were abrogated in the presence of receptor-associated protein-1, an antagonist of the low-density lipoprotein receptor family, which also inhibited MT- and EmtinB-induced neurite outgrowth and survival. MT-mediated neurite outgrowth was furthermore inhibited by an anti-megalin serum. EmtinB-mediated inhibition of apoptosis occurred without a reduction of caspase-3 activity, but was associated with reduced expression of the pro-apoptotic B-cell leukemia/lymphoma-2 interacting member of cell death (Bim(S)). Finally, evidence is provided that MT and EmtinB activate extracellular signal-regulated kinase, protein kinase B, and cAMP response element binding protein. Altogether, these results strongly suggest that MT and EmtinB induce their neuronal effects through direct binding to surface receptors belonging to the low-density lipoprotein receptor family, such as megalin and LRP, thereby activating signal transduction pathways resulting in neurite outgrowth and survival.


Subject(s)
Cell Differentiation/drug effects , Metallothionein/pharmacology , Neurons/drug effects , Peptides/pharmacology , Receptors, LDL/physiology , Analysis of Variance , Animals , Animals, Newborn , Cell Survival/drug effects , Cells, Cultured , Cerebellum/cytology , Dose-Response Relationship, Drug , In Situ Nick-End Labeling/methods , Intercellular Signaling Peptides and Proteins , LDL-Receptor Related Protein-Associated Protein/metabolism , Low Density Lipoprotein Receptor-Related Protein-2/metabolism , Metallothionein/chemistry , Models, Biological , Neurites/drug effects , Peptides/chemistry , Protein Binding/drug effects , Rats , Rats, Wistar , Signal Transduction/physiology , Surface Plasmon Resonance/methods
10.
J Invest Dermatol ; 124(1): 151-9, 2005 Jan.
Article in English | MEDLINE | ID: mdl-15654969

ABSTRACT

We have characterized a novel animal model of the common inflammatory skin disease seborrheic dermatitis (SD) that involves the expression of the self-specific 2C transgenic T cell receptor on the DBA/2 genetic background. Opportunistic fungal pathogens are present in the primary histological lesions and severe disease can be mitigated by the administration of fluconazole, demonstrating a role for infection in disease pathogenesis. Spontaneous disease convalescence occurs at 70-90 d of age and is preceded by an expansion of CD4+ T cells that partially restores the T cell lymphopenia that occurs in these animals. The adoptive transfer of syngeneic CD4+ T cells into pre-diseased DBA/2 2C mice completely abrogates the development of cutaneous disease. The pattern of disease inheritance in DBA/2 backcrosses suggests that one, or a closely linked group of genes, may control disease penetrance. Bone marrow reconstitution experiments demonstrated that the DBA/2 susceptibility factor(s) governing disease penetrance is likely non-hematopoietic since bone marrow from disease-resistant 2C mice can adoptively transfer the full disease phenotype to non-transgenic DBA/2 animals. This model implicates fungal organisms and CD4+ T cell lymphopenia in the development of a SD-like condition and, as such, may mimic the development of SD in acquired immunodeficiency syndrome.


Subject(s)
Dermatitis, Seborrheic/genetics , Dermatitis, Seborrheic/immunology , Disease Models, Animal , Mice, Transgenic , Receptors, Antigen, T-Cell/genetics , Adoptive Transfer , Age Factors , Animals , Antigens/immunology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/transplantation , Dermatitis, Seborrheic/pathology , Genetic Predisposition to Disease , Immunocompromised Host/genetics , Immunocompromised Host/immunology , Lymphopenia/genetics , Lymphopenia/immunology , Lymphopenia/pathology , Mice , Mice, Inbred BALB C , Mice, Inbred DBA , Mycoses/immunology , Mycoses/pathology , Receptors, Antigen, T-Cell/immunology , Sexual Maturation
SELECTION OF CITATIONS
SEARCH DETAIL
...