Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 40
Filter
1.
EMBO J ; 41(4): e109108, 2022 02 15.
Article in English | MEDLINE | ID: mdl-35019161

ABSTRACT

Haploinsufficiency of the progranulin (PGRN)-encoding gene (GRN) causes frontotemporal lobar degeneration (GRN-FTLD) and results in microglial hyperactivation, TREM2 activation, lysosomal dysfunction, and TDP-43 deposition. To understand the contribution of microglial hyperactivation to pathology, we used genetic and pharmacological approaches to suppress TREM2-dependent transition of microglia from a homeostatic to a disease-associated state. Trem2 deficiency in Grn KO mice reduced microglia hyperactivation. To explore antibody-mediated pharmacological modulation of TREM2-dependent microglial states, we identified antagonistic TREM2 antibodies. Treatment of macrophages from GRN-FTLD patients with these antibodies led to reduced TREM2 signaling due to its enhanced shedding. Furthermore, TREM2 antibody-treated PGRN-deficient microglia derived from human-induced pluripotent stem cells showed reduced microglial hyperactivation, TREM2 signaling, and phagocytic activity, but lysosomal dysfunction was not rescued. Similarly, lysosomal dysfunction, lipid dysregulation, and glucose hypometabolism of Grn KO mice were not rescued by TREM2 ablation. Synaptic loss and neurofilament light-chain (NfL) levels, a biomarker for neurodegeneration, were further elevated in the Grn/Trem2 KO cerebrospinal fluid (CSF). These findings suggest that TREM2-dependent microglia hyperactivation in models of GRN deficiency does not promote neurotoxicity, but rather neuroprotection.


Subject(s)
Frontotemporal Lobar Degeneration/pathology , Membrane Glycoproteins/metabolism , Microglia/physiology , Monocytes/metabolism , Progranulins/deficiency , Receptors, Immunologic/metabolism , Animals , Antibodies/immunology , Antibodies/pharmacology , Brain/diagnostic imaging , Brain/physiopathology , Disease Models, Animal , Female , Frontotemporal Lobar Degeneration/metabolism , Humans , Lysosomes/metabolism , Lysosomes/pathology , Male , Membrane Glycoproteins/genetics , Membrane Glycoproteins/immunology , Mice, Inbred C57BL , Mice, Knockout , Microglia/drug effects , Monocytes/drug effects , Receptors, Immunologic/genetics , Receptors, Immunologic/immunology , Syk Kinase/metabolism
2.
J Neuroinflammation ; 20(1): 47, 2023 Feb 24.
Article in English | MEDLINE | ID: mdl-36829182

ABSTRACT

AIM: We aimed to investigate the impact of microglial activity and microglial FDG uptake on metabolic connectivity, since microglial activation states determine FDG-PET alterations. Metabolic connectivity refers to a concept of interacting metabolic brain regions and receives growing interest in approaching complex cerebral metabolic networks in neurodegenerative diseases. However, underlying sources of metabolic connectivity remain to be elucidated. MATERIALS AND METHODS: We analyzed metabolic networks measured by interregional correlation coefficients (ICCs) of FDG-PET scans in WT mice and in mice with mutations in progranulin (Grn) or triggering receptor expressed on myeloid cells 2 (Trem2) knockouts (-/-) as well as in double mutant Grn-/-/Trem2-/- mice. We selected those rodent models as they represent opposite microglial signatures with disease associated microglia in Grn-/- mice and microglia locked in a homeostatic state in Trem2-/- mice; however, both resulting in lower glucose uptake of the brain. The direct influence of microglia on metabolic networks was further determined by microglia depletion using a CSF1R inhibitor in WT mice at two different ages. Within maps of global mean scaled regional FDG uptake, 24 pre-established volumes of interest were applied and assigned to either cortical or subcortical networks. ICCs of all region pairs were calculated and z-transformed prior to group comparisons. FDG uptake of neurons, microglia, and astrocytes was determined in Grn-/- and WT mice via assessment of single cell tracer uptake (scRadiotracing). RESULTS: Microglia depletion by CSF1R inhibition resulted in a strong decrease of metabolic connectivity defined by decrease of mean cortical ICCs in WT mice at both ages studied (6-7 m; p = 0.0148, 9-10 m; p = 0.0191), when compared to vehicle-treated age-matched WT mice. Grn-/-, Trem2-/- and Grn-/-/Trem2-/- mice all displayed reduced FDG-PET signals when compared to WT mice. However, when analyzing metabolic networks, a distinct increase of ICCs was observed in Grn-/- mice when compared to WT mice in cortical (p < 0.0001) and hippocampal (p < 0.0001) networks. In contrast, Trem2-/- mice did not show significant alterations in metabolic connectivity when compared to WT. Furthermore, the increased metabolic connectivity in Grn-/- mice was completely suppressed in Grn-/-/Trem2-/- mice. Grn-/- mice exhibited a severe loss of neuronal FDG uptake (- 61%, p < 0.0001) which shifted allocation of cellular brain FDG uptake to microglia (42% in Grn-/- vs. 22% in WT). CONCLUSIONS: Presence, absence, and activation of microglia have a strong impact on metabolic connectivity of the mouse brain. Enhanced metabolic connectivity is associated with increased microglial FDG allocation.


Subject(s)
Fluorodeoxyglucose F18 , Microglia , Animals , Mice , Microglia/metabolism , Fluorodeoxyglucose F18/metabolism , Progranulins/metabolism , Brain/metabolism , Positron-Emission Tomography , Membrane Glycoproteins/metabolism , Receptors, Immunologic/metabolism
3.
EMBO Rep ; 21(10): e50241, 2020 10 05.
Article in English | MEDLINE | ID: mdl-32929860

ABSTRACT

Single nucleotide polymorphisms (SNPs) in TMEM106B encoding the lysosomal type II transmembrane protein 106B increase the risk for frontotemporal lobar degeneration (FTLD) of GRN (progranulin gene) mutation carriers. Currently, it is unclear if progranulin (PGRN) and TMEM106B are synergistically linked and if a gain or a loss of function of TMEM106B is responsible for the increased disease risk of patients with GRN haploinsufficiency. We therefore compare behavioral abnormalities, gene expression patterns, lysosomal activity, and TDP-43 pathology in single and double knockout animals. Grn-/- /Tmem106b-/- mice show a strongly reduced life span and massive motor deficits. Gene expression analysis reveals an upregulation of molecular signature characteristic for disease-associated microglia and autophagy. Dysregulation of maturation of lysosomal proteins as well as an accumulation of ubiquitinated proteins and widespread p62 deposition suggest that proteostasis is impaired. Moreover, while single Grn-/- knockouts only occasionally show TDP-43 pathology, the double knockout mice exhibit deposition of phosphorylated TDP-43. Thus, a loss of function of TMEM106B may enhance the risk for GRN-associated FTLD by reduced protein turnover in the lysosomal/autophagic system.


Subject(s)
Frontotemporal Lobar Degeneration , Intercellular Signaling Peptides and Proteins , Animals , Frontotemporal Lobar Degeneration/genetics , Humans , Intercellular Signaling Peptides and Proteins/genetics , Lysosomes , Membrane Proteins/genetics , Mice , Mice, Knockout , Nerve Tissue Proteins , Progranulins/genetics
4.
EMBO Rep ; 18(7): 1186-1198, 2017 07.
Article in English | MEDLINE | ID: mdl-28483841

ABSTRACT

Sequence variations in the triggering receptor expressed on myeloid cells 2 (TREM2) have been linked to an increased risk for neurodegenerative disorders such as Alzheimer's disease and frontotemporal lobar degeneration. In the brain, TREM2 is predominantly expressed in microglia. Several disease-associated TREM2 variants result in a loss of function by reducing microglial phagocytosis, impairing lipid sensing, preventing binding of lipoproteins and affecting shielding of amyloid plaques. We here investigate the consequences of TREM2 loss of function on the microglia transcriptome. Among the differentially expressed messenger RNAs in wild-type and Trem2-/- microglia, gene clusters are identified which represent gene functions in chemotaxis, migration and mobility. Functional analyses confirm that loss of TREM2 impairs appropriate microglial responses to injury and signals that normally evoke chemotaxis on multiple levels. In an ex vivo organotypic brain slice assay, absence of TREM2 reduces the distance migrated by microglia. Moreover, migration towards defined chemo-attractants is reduced upon ablation of TREM2 and can be rescued by TREM2 re-expression. In vivo, microglia lacking TREM2 migrate less towards injected apoptotic neurons, and outgrowth of microglial processes towards sites of laser-induced focal CNS damage in the somatosensory cortex is slowed. The apparent lack of chemotactic stimulation upon depletion of TREM2 is consistent with a stable expression profile of genes characterizing the homoeostatic signature of microglia.


Subject(s)
Chemotaxis , Membrane Glycoproteins/deficiency , Membrane Glycoproteins/genetics , Microglia/physiology , Neurons/pathology , Receptors, Immunologic/deficiency , Receptors, Immunologic/genetics , Alzheimer Disease/genetics , Alzheimer Disease/physiopathology , Cells, Cultured , Frontotemporal Dementia , Gene Expression Profiling , Humans , Loss of Function Mutation , Myeloid Cells , Neurodegenerative Diseases/genetics , Neurodegenerative Diseases/physiopathology , Phagocytosis
5.
EMBO J ; 33(5): 450-67, 2014 Mar 03.
Article in English | MEDLINE | ID: mdl-24357581

ABSTRACT

TMEM106B is a major risk factor for frontotemporal lobar degeneration with TDP-43 pathology. TMEM106B localizes to lysosomes, but its function remains unclear. We show that TMEM106B knockdown in primary neurons affects lysosomal trafficking and blunts dendritic arborization. We identify microtubule-associated protein 6 (MAP6) as novel interacting protein for TMEM106B. MAP6 over-expression inhibits dendritic branching similar to TMEM106B knockdown. MAP6 knockdown fully rescues the dendritic phenotype of TMEM106B knockdown, supporting a functional interaction between TMEM106B and MAP6. Live imaging reveals that TMEM106B knockdown and MAP6 overexpression strongly increase retrograde transport of lysosomes in dendrites. Downregulation of MAP6 in TMEM106B knockdown neurons restores the balance of anterograde and retrograde lysosomal transport and thereby prevents loss of dendrites. To strengthen the link, we enhanced anterograde lysosomal transport by expressing dominant-negative Rab7-interacting lysosomal protein (RILP), which also rescues the dendrite loss in TMEM106B knockdown neurons. Thus, TMEM106B/MAP6 interaction is crucial for controlling dendritic trafficking of lysosomes, presumably by acting as a molecular brake for retrograde transport. Lysosomal misrouting may promote neurodegeneration in patients with TMEM106B risk variants.


Subject(s)
Dendrites/metabolism , Lysosomes/metabolism , Membrane Proteins/metabolism , Microtubule-Associated Proteins/metabolism , Nerve Tissue Proteins/metabolism , Animals , Cell Line , Gene Expression , Gene Knockdown Techniques , Humans , Protein Interaction Mapping , Rats
6.
Genet Med ; 20(2): 240-249, 2018 02.
Article in English | MEDLINE | ID: mdl-28749476

ABSTRACT

PurposeTo define the genetic spectrum and relative gene frequencies underlying clinical frontotemporal dementia (FTD).MethodsWe investigated the frequencies and mutations in neurodegenerative disease genes in 121 consecutive FTD subjects using an unbiased, combined sequencing approach, complemented by cerebrospinal fluid Aß1-42 and serum progranulin measurements. Subjects were screened for C9orf72 repeat expansions, GRN and MAPT mutations, and, if negative, mutations in other neurodegenerative disease genes, by whole-exome sequencing (WES) (n = 108), including WES-based copy-number variant (CNV) analysis.ResultsPathogenic and likely pathogenic mutations were identified in 19% of the subjects, including mutations in C9orf72 (n = 8), GRN (n = 7, one 11-exon macro-deletion) and, more rarely, CHCHD10, TARDBP, SQSTM1 and UBQLN2 (each n = 1), but not in MAPT or TBK1. WES also unraveled pathogenic mutations in genes not commonly linked to FTD, including mutations in Alzheimer (PSEN1, PSEN2), lysosomal (CTSF, 7-exon macro-deletion) and cholesterol homeostasis pathways (CYP27A1).ConclusionOur unbiased approach reveals a wide genetic spectrum underlying clinical FTD, including 11% of seemingly sporadic FTD. It unravels several mutations and CNVs in genes and pathways hitherto not linked to FTD. This suggests that clinical FTD might be the converging downstream result of a delicate susceptibility of frontotemporal brain networks to insults in various pathways.


Subject(s)
Frontotemporal Dementia/epidemiology , Frontotemporal Dementia/genetics , Genetic Association Studies , Genetic Predisposition to Disease , Alleles , Biomarkers , C9orf72 Protein/genetics , Female , Frontotemporal Dementia/diagnosis , Frontotemporal Dementia/metabolism , Gene Frequency , Genetic Association Studies/methods , Genetic Testing , Genotype , Humans , Magnetic Resonance Imaging , Male , Middle Aged , Mutation , Neurodegenerative Diseases/genetics , Pedigree , Phenotype , Sequence Analysis, DNA , Exome Sequencing
7.
J Biol Chem ; 289(37): 25879-89, 2014 Sep 12.
Article in English | MEDLINE | ID: mdl-25056957

ABSTRACT

Frontotemporal lobar degeneration is associated with cytoplasmic or nuclear deposition of the TAR DNA-binding protein 43 (TDP-43). Haploinsufficiency of progranulin (GRN) is a major genetic risk factor for frontotemporal lobar degeneration associated with TDP-43 deposition. Therefore, understanding the mechanisms that control cellular expression of GRN is required not only to understand disease etiology but also for the development of potential therapeutic strategies. We identified different GRN transcripts with short (38-93 nucleotides) or long (219 nucleotides) 5' UTRs and demonstrate a cellular mechanism that represses translation of GRN mRNAs with long 5' UTRs. The long 5' UTR of GRN mRNA contains an upstream open reading frame (uORF) that is absent in all shorter transcripts. Because such UTRs can be involved in translational control as well as in mRNA stability, we compared the expression of GRN in cells expressing cDNAs with and without 5' UTRs. This revealed a selective repression of GRN translation and a reduction of mRNA levels by the 219-nucleotide-long 5' UTR. The specific ability of this GRN 5' UTR to repress protein expression was further confirmed by its transfer to an independent reporter. Deletion analysis identified a short stretch between nucleotides 76 and 125 containing two start codons within one uORF that is required and sufficient for repression of protein expression. Mutagenesis of the two AUG codons within the uORF is sufficient to reduce translational repression. Therefore initiating ribosomes at the AUGs of the uORF fail to efficiently initiate translation at the start codon of GRN. In parallel the 5' UTR also affects mRNA stability; thus two independent mechanisms determine GRN expression via mRNA stability and translational efficiency.


Subject(s)
Amyotrophic Lateral Sclerosis/genetics , DNA-Binding Proteins/genetics , Frontotemporal Lobar Degeneration/metabolism , Intercellular Signaling Peptides and Proteins/genetics , Peptide Chain Termination, Translational/genetics , 3' Untranslated Regions/genetics , 5' Untranslated Regions/genetics , Amyotrophic Lateral Sclerosis/metabolism , Amyotrophic Lateral Sclerosis/pathology , DNA-Binding Proteins/metabolism , Frontotemporal Lobar Degeneration/genetics , Frontotemporal Lobar Degeneration/pathology , Gene Expression Regulation , HEK293 Cells , Humans , Intercellular Signaling Peptides and Proteins/metabolism , Mutation , Open Reading Frames , Progranulins , Protein Isoforms/genetics , RNA, Messenger/genetics
8.
Traffic ; 13(10): 1393-410, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22758778

ABSTRACT

ß-Amyloid (Aß) peptides are generated from the successive proteolytic processing of the amyloid precursor protein (APP) by the ß-APP cleaving enzyme (BACE or ß-secretase) and the γ-secretase complex. Initial cleavage of APP by BACE leads into the amyloidogenic pathway, causing or exacerbating Alzheimer's disease. Therefore, their intracellular traffic can determine how easily and frequently BACE has access to and cleaves APP. Here, we have used polarized Madin-Darby canine kidney (MDCK) cells stably expressing APP and BACE to examine the regulation of their polarized trafficking by retromer, a protein complex previously implicated in their endosome-to-Golgi transport. Our data show that retromer interacts with BACE and regulates its postendocytic sorting in polarized MDCK cells. Depleting retromer, inhibiting retromer function, or preventing BACE interaction with retromer, alters trafficking of BACE, which thereby increases its localization in the early endocytic compartment. As a result, this slows endocytosis of apically localized BACE, promoting its recycling and apical-to-basolateral transcytosis, which increases APP/BACE interaction and subsequent cleavage of APP toward generation and secretion of Aß peptides.


Subject(s)
Amyloid Precursor Protein Secretases/metabolism , Endocytosis , Vesicular Transport Proteins/metabolism , Amyloid Precursor Protein Secretases/chemistry , Amyloid Precursor Protein Secretases/genetics , Amyloid beta-Protein Precursor/metabolism , Animals , Cell Line , Dogs , Endosomes/metabolism , Golgi Apparatus/metabolism , Madin Darby Canine Kidney Cells , Mice , Multiprotein Complexes/metabolism , Mutation , Protein Interaction Domains and Motifs , Protein Transport
9.
EMBO J ; 29(16): 2841-57, 2010 Aug 18.
Article in English | MEDLINE | ID: mdl-20606625

ABSTRACT

Mutations in fused in sarcoma (FUS) are a cause of familial amyotrophic lateral sclerosis (fALS). Patients carrying point mutations in the C-terminus of FUS show neuronal cytoplasmic FUS-positive inclusions, whereas in healthy controls, FUS is predominantly nuclear. Cytoplasmic FUS inclusions have also been identified in a subset of frontotemporal lobar degeneration (FTLD-FUS). We show that a non-classical PY nuclear localization signal (NLS) in the C-terminus of FUS is necessary for nuclear import. The majority of fALS-associated mutations occur within the NLS and impair nuclear import to a degree that correlates with the age of disease onset. This presents the first case of disease-causing mutations within a PY-NLS. Nuclear import of FUS is dependent on Transportin, and interference with this transport pathway leads to cytoplasmic redistribution and recruitment of FUS into stress granules. Moreover, proteins known to be stress granule markers co-deposit with inclusions in fALS and FTLD-FUS patients, implicating stress granule formation in the pathogenesis of these diseases. We propose that two pathological hits, namely nuclear import defects and cellular stress, are involved in the pathogenesis of FUS-opathies.


Subject(s)
Amyotrophic Lateral Sclerosis/genetics , Karyopherins/metabolism , Point Mutation , RNA-Binding Protein FUS/genetics , RNA-Binding Protein FUS/metabolism , Active Transport, Cell Nucleus , Amino Acid Sequence , Amyotrophic Lateral Sclerosis/metabolism , Amyotrophic Lateral Sclerosis/pathology , Animals , Cells, Cultured , Cytoplasmic Granules/pathology , Gene Knockdown Techniques , HeLa Cells , Humans , Karyopherins/genetics , Molecular Sequence Data , Neurons/pathology , Protein Structure, Tertiary , RNA-Binding Protein FUS/analysis , RNA-Binding Protein FUS/chemistry , Zebrafish/embryology
10.
Acta Neuropathol ; 127(6): 845-60, 2014.
Article in English | MEDLINE | ID: mdl-24619111

ABSTRACT

Heterozygous loss-of-function mutations in the progranulin (GRN) gene and the resulting reduction of GRN levels is a common genetic cause for frontotemporal lobar degeneration (FTLD) with accumulation of TAR DNA-binding protein (TDP)-43. Recently, it has been shown that a complete GRN deficiency due to a homozygous GRN loss-of-function mutation causes neuronal ceroid lipofuscinosis (NCL), a lysosomal storage disorder. These findings suggest that lysosomal dysfunction may also contribute to some extent to FTLD. Indeed, Grn(-/-) mice recapitulate not only pathobiochemical features of GRN-associated FTLD-TDP (FTLD-TDP/GRN), but also those which are characteristic for NCL and lysosomal impairment. In Grn(-/-) mice the lysosomal proteins cathepsin D (CTSD), LAMP (lysosomal-associated membrane protein) 1 and the NCL storage components saposin D and subunit c of mitochondrial ATP synthase (SCMAS) were all found to be elevated. Moreover, these mice display increased levels of transmembrane protein (TMEM) 106B, a lysosomal protein known as a risk factor for FTLD-TDP pathology. In line with a potential pathological overlap of FTLD and NCL, Ctsd(-/-) mice, a model for NCL, show elevated levels of the FTLD-associated proteins GRN and TMEM106B. In addition, pathologically phosphorylated TDP-43 occurs in Ctsd(-/-) mice to a similar extent as in Grn(-/-) mice. Consistent with these findings, some NCL patients accumulate pathologically phosphorylated TDP-43 within their brains. Based on these observations, we searched for pathological marker proteins, which are characteristic for NCL or lysosomal impairment in brains of FTLD-TDP/GRN patients. Strikingly, saposin D, SCMAS as well as the lysosomal proteins CTSD and LAMP1/2 are all elevated in patients with FTLD-TDP/GRN. Thus, our findings suggest that lysosomal storage disorders and GRN-associated FTLD may share common features.


Subject(s)
Brain/metabolism , Brain/pathology , Frontotemporal Lobar Degeneration/metabolism , Frontotemporal Lobar Degeneration/pathology , Neuronal Ceroid-Lipofuscinoses/metabolism , Neuronal Ceroid-Lipofuscinoses/pathology , Animals , Cathepsin D/genetics , Cathepsin D/metabolism , DNA-Binding Proteins/metabolism , Disease Models, Animal , Granulins , Humans , Immunoblotting , Immunohistochemistry , Intercellular Signaling Peptides and Proteins/deficiency , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/metabolism , Mice, Knockout , Phosphorylation , Progranulins
11.
Sci Transl Med ; 16(750): eadj7308, 2024 Jun 05.
Article in English | MEDLINE | ID: mdl-38838131

ABSTRACT

Progranulin (PGRN) haploinsufficiency is a major risk factor for frontotemporal lobar degeneration with TAR DNA-binding protein 43 (TDP-43) pathology (FTLD-GRN). Multiple therapeutic strategies are in clinical development to restore PGRN in the CNS, including gene therapy. However, a limitation of current gene therapy approaches aimed to alleviate FTLD-associated pathologies may be their inefficient brain exposure and biodistribution. We therefore developed an adeno-associated virus (AAV) targeting the liver (L) to achieve sustained peripheral expression of a transferrin receptor (TfR) binding, brain-penetrant (b) PGRN variant [AAV(L):bPGRN] in two mouse models of FTLD-GRN, namely, Grn knockout and GrnxTmem106b double knockout mice. This therapeutic strategy avoids potential safety and biodistribution issues of CNS-administered AAVs and maintains sustained concentrations of PGRN in the brain after a single dose. AAV(L):bPGRN treatment reduced several FTLD-GRN-associated pathologies including severe motor function deficits, aberrant TDP-43 phosphorylation, dysfunctional protein degradation, lipid metabolism, gliosis, and neurodegeneration in the brain. The potential translatability of our findings was tested in an in vitro model using cocultured human induced pluripotent stem cell (hiPSC)-derived microglia lacking PGRN and TMEM106B and wild-type hiPSC-derived neurons. As in mice, aberrant TDP-43, lysosomal dysfunction, and neuronal loss were ameliorated after treatment with exogenous TfR-binding protein transport vehicle fused to PGRN (PTV:PGRN). Together, our studies suggest that peripherally administered brain-penetrant PGRN replacement strategies ameliorate FTLD-GRN relevant phenotypes including TDP-43 pathology, neurodegeneration, and behavioral deficits. Our data provide preclinical proof of concept for the use of this AAV platform for treatment of FTLD-GRN and potentially other CNS disorders.


Subject(s)
Brain , Dependovirus , Disease Models, Animal , Frontotemporal Lobar Degeneration , Mice, Knockout , Progranulins , Animals , Humans , Mice , Brain/metabolism , Brain/pathology , Dependovirus/metabolism , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/genetics , Frontotemporal Lobar Degeneration/metabolism , Frontotemporal Lobar Degeneration/pathology , Genetic Therapy , Phosphorylation , Progranulins/metabolism , Progranulins/genetics , Receptors, Transferrin/metabolism
12.
J Biol Chem ; 287(23): 19355-65, 2012 Jun 01.
Article in English | MEDLINE | ID: mdl-22511793

ABSTRACT

TMEM106B was identified as a major risk factor in a genome-wide association study for frontotemporal lobar degeneration (FTLD) with TAR DNA-binding protein (TDP)-43 pathology. The most significant association of TMEM106B single nucleotide polymorphisms with risk of FTLD-TDP was observed in patients with progranulin (GRN) mutations. Subsequent studies suggested an inverse correlation between TMEM106B expression and GRN levels in patient serum. However, in this study, this was not confirmed as we failed to detect a significant alteration of GRN levels upon knockdown or exogenous expression of TMEM106B in heterologous cells. To provide a basis for understanding TMEM106B function in health and disease, we investigated the membrane orientation and subcellular localization of this completely uncharacterized protein. By differential membrane extraction and sequential mutagenesis of potential N-glycosylation sites, we identified TMEM106B as a type 2 integral membrane protein with a highly glycosylated luminal domain. Glycosylation is partially required for the transport of TMEM106B beyond the endoplasmic reticulum to late cellular compartments. Endogenous as well as overexpressed TMEM106B localizes to late endosomes and lysosomes. Interestingly, the inhibition of vacuolar H(+)-ATPases significantly increased the levels of TMEM106B, a finding that may provide an unexpected biochemical link to GRN, because this protein is also strongly increased under the same conditions. Our findings provide a biochemical and cell biological basis for the understanding of the pathological role of TMEM106B in FTLD, an incurable neurodegenerative disorder.


Subject(s)
Cell Membrane/metabolism , Endosomes/metabolism , Frontotemporal Lobar Degeneration/metabolism , Lysosomes/metabolism , Membrane Proteins/metabolism , Nerve Tissue Proteins/metabolism , Polymorphism, Single Nucleotide , Cell Membrane/genetics , Cell Membrane/pathology , Endosomes/genetics , Endosomes/pathology , Frontotemporal Lobar Degeneration/genetics , Frontotemporal Lobar Degeneration/pathology , Genome-Wide Association Study , Glycosylation , HEK293 Cells , HeLa Cells , Humans , Lysosomes/genetics , Lysosomes/pathology , Membrane Proteins/genetics , Nerve Tissue Proteins/genetics , Protein Transport/genetics , Risk Factors , Vacuolar Proton-Translocating ATPases/genetics , Vacuolar Proton-Translocating ATPases/metabolism
13.
J Pathol ; 228(1): 67-76, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22733568

ABSTRACT

Loss-of-function mutations in progranulin (GRN) are associated with frontotemporal lobar degeneration with intraneuronal ubiquitinated protein accumulations composed primarily of hyperphosphorylated TDP-43 (FTLD-TDP). The mechanism by which GRN deficiency causes TDP-43 pathology or neurodegeneration remains elusive. To explore the role of GRN in vivo, we established Grn knockout mice using a targeted genomic recombination approach and Cre-LoxP technology. Constitutive Grn homozygous knockout (Grn(-/-) ) mice were born in an expected Mendelian pattern of inheritance and showed no phenotypic alterations compared to heterozygous (Grn(+/-) ) or wild-type (Wt) littermates until 10 months of age. From then, Grn(-/-) mice showed reduced survival accompanied by significantly increased gliosis and ubiquitin-positive accumulations in the cortex, hippocampus, and subcortical regions. Although phosphorylated TDP-43 could not be detected in the ubiquitinated inclusions, elevated levels of hyperphosphorylated full-length TDP-43 were recovered from detergent-insoluble brain fractions of Grn(-/-) mice. Phosphorylated TDP-43 increased with age and was primarily extracted from the nuclear fraction. Grn(-/-) mice also showed degenerative liver changes and cathepsin D-positive foamy histiocytes within sinusoids, suggesting widespread defects in lysosomal turnover. An increase in insulin-like growth factor (IGF)-1 was observed in Grn(-/-) brains, and increased IGF-1 signalling has been associated with decreased longevity. Our data suggest that progranulin deficiency in mice leads to reduced survival in adulthood and increased cellular ageing accompanied by hyperphosphorylation of TDP-43, and recapitulates key aspects of FTLD-TDP neuropathology.


Subject(s)
Cellular Senescence , DNA-Binding Proteins/metabolism , Frontotemporal Lobar Degeneration/pathology , Intercellular Signaling Peptides and Proteins/physiology , Animals , Behavior, Animal/physiology , Brain/metabolism , Brain/pathology , DNA-Binding Proteins/genetics , Female , Frontotemporal Lobar Degeneration/genetics , Frontotemporal Lobar Degeneration/mortality , Gene Expression , Gliosis/metabolism , Gliosis/pathology , Granulins , Liver/pathology , Longevity/physiology , Male , Mice , Mice, Knockout , Phosphorylation , Progranulins , Survival Rate , Ubiquitin/metabolism
14.
Acta Neuropathol Commun ; 11(1): 21, 2023 01 27.
Article in English | MEDLINE | ID: mdl-36707901

ABSTRACT

Genetic variants in TMEM106B are a common risk factor for frontotemporal lobar degeneration and the most important modifier of disease risk in patients with progranulin (GRN) mutations (FTLD-GRN). TMEM106B is encoding a lysosomal transmembrane protein of unknown molecular function. How it mediates its disease-modifying function remains enigmatic. Several TMEM106B single nucleotide polymorphisms (SNPs) are significantly associated with disease risk in FTLD-GRN carriers, of which all except one are within intronic sequences of TMEM106B. Of note, the non-coding SNPs are in high linkage disequilibrium with the coding SNP rs3173615 located in exon six of TMEM106B, resulting in a threonine to serine change at amino acid 185 in the minor allele, which is protective in FTLD-GRN carriers. To investigate the functional consequences of this variant in vivo, we generated and characterized a knockin mouse model harboring the Tmem106bT186S variant. We analyzed the effect of this protective variant on FTLD pathology by crossing Tmem106bT186S mice with Grn-/- knockout mice, a model for GRN-mediated FTLD. We did not observe the amelioration of any of the investigated Grn-/- knockout phenotypes, including transcriptomic changes, lipid alterations, or microgliosis in Tmem106bT186S/T186S × Grn-/- mice, indicating that the Tmem106bT186S variant is not protective in the Grn-/- knockout mouse model. These data suggest that effects of the associated SNPs not directly linked to the amino acid exchange in TMEM106B are critical for the modifying effect.


Subject(s)
Frontotemporal Dementia , Frontotemporal Lobar Degeneration , Animals , Mice , Amino Acids , Frontotemporal Dementia/genetics , Frontotemporal Lobar Degeneration/pathology , Intercellular Signaling Peptides and Proteins/genetics , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice, Knockout , Mutation , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Polymorphism, Single Nucleotide/genetics
15.
Nat Neurosci ; 26(3): 416-429, 2023 03.
Article in English | MEDLINE | ID: mdl-36635496

ABSTRACT

Loss-of-function variants of TREM2 are associated with increased risk of Alzheimer's disease (AD), suggesting that activation of this innate immune receptor may be a useful therapeutic strategy. Here we describe a high-affinity human TREM2-activating antibody engineered with a monovalent transferrin receptor (TfR) binding site, termed antibody transport vehicle (ATV), to facilitate blood-brain barrier transcytosis. Upon peripheral delivery in mice, ATV:TREM2 showed improved brain biodistribution and enhanced signaling compared to a standard anti-TREM2 antibody. In human induced pluripotent stem cell (iPSC)-derived microglia, ATV:TREM2 induced proliferation and improved mitochondrial metabolism. Single-cell RNA sequencing and morphometry revealed that ATV:TREM2 shifted microglia to metabolically responsive states, which were distinct from those induced by amyloid pathology. In an AD mouse model, ATV:TREM2 boosted brain microglial activity and glucose metabolism. Thus, ATV:TREM2 represents a promising approach to improve microglial function and treat brain hypometabolism found in patients with AD.


Subject(s)
Alzheimer Disease , Induced Pluripotent Stem Cells , Humans , Animals , Mice , Microglia , Blood-Brain Barrier , Tissue Distribution , Antibodies , Brain , Disease Models, Animal , Membrane Glycoproteins , Receptors, Immunologic/genetics
16.
Traffic ; 11(2): 250-8, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19958468

ABSTRACT

gamma-Secretase is critically involved in the Notch pathway and in Alzheimer's disease. The four subunits of gamma-secretase assemble in the endoplasmic reticulum (ER) and unassembled subunits are retained/retrieved to the ER by specific signals. We here describe a novel ER-retention/retrieval signal in the transmembrane domain (TMD) 4 of presenilin 1, a subunit of gamma-secretase. TMD4 also is essential for complex formation, conferring a dual role for this domain. Likewise, TMD1 of Pen2 is bifunctional as well. It carries an ER-retention/retrieval signal and is important for complex assembly by binding to TMD4. The two TMDs directly interact with each other and mask their respective ER-retention/retrieval signals, allowing surface transport of reporter proteins. Our data suggest a model how assembly of Pen2 into the nascent gamma-secretase complex could mask TMD-based ER-retention/retrieval signals to allow plasma membrane transport of fully assembled gamma-secretase.


Subject(s)
Amyloid Precursor Protein Secretases/metabolism , Endoplasmic Reticulum/metabolism , Protein Sorting Signals , Amyloid Precursor Protein Secretases/chemistry , Amyloid Precursor Protein Secretases/genetics , Animals , Cell Line , Humans , Immunoblotting , Mice , Microscopy, Fluorescence , Presenilins/chemistry , Presenilins/genetics , Protein Binding , Protein Structure, Tertiary , Protein Transport
17.
J Neurosci ; 31(5): 1885-94, 2011 Feb 02.
Article in English | MEDLINE | ID: mdl-21289198

ABSTRACT

Numerous loss-of-function mutations in the progranulin (GRN) gene cause frontotemporal lobar degeneration with ubiquitin and TAR-DNA binding protein 43-positive inclusions by reduced production and secretion of GRN. Consistent with the observation that GRN has neurotrophic properties, pharmacological stimulation of GRN production is a promising approach to rescue GRN haploinsufficiency and prevent disease progression. We therefore searched for compounds capable of selectively increasing GRN levels. Here, we demonstrate that four independent and highly selective inhibitors of vacuolar ATPase (bafilomycin A1, concanamycin A, archazolid B, and apicularen A) significantly elevate intracellular and secreted GRN. Furthermore, clinically used alkalizing drugs, including chloroquine, bepridil, and amiodarone, similarly stimulate GRN production. Elevation of GRN levels occurs via a translational mechanism independent of lysosomal degradation, autophagy, or endocytosis. Importantly, alkalizing reagents rescue GRN deficiency in organotypic cortical slice cultures from a mouse model for GRN deficiency and in primary cells derived from human patients with GRN loss-of-function mutations. Thus, alkalizing reagents, specifically those already used in humans for other applications, and vacuolar ATPase inhibitors may be therapeutically used to prevent GRN-dependent neurodegeneration.


Subject(s)
Alkalies/pharmacology , Cerebral Cortex/metabolism , Fibroblasts/metabolism , Frontotemporal Lobar Degeneration/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Neurons/metabolism , Vacuolar Proton-Translocating ATPases/antagonists & inhibitors , Amiodarone/pharmacology , Animals , Animals, Newborn , Autophagy-Related Protein 5 , Bepridil/pharmacology , Blotting, Northern , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Cells, Cultured , Cerebral Cortex/drug effects , Chloroquine/pharmacology , Enzyme-Linked Immunosorbent Assay , Female , Fibroblasts/drug effects , Frontotemporal Lobar Degeneration/drug therapy , Frontotemporal Lobar Degeneration/genetics , Granulins , HEK293 Cells , HeLa Cells , Humans , Intercellular Signaling Peptides and Proteins/deficiency , Intercellular Signaling Peptides and Proteins/genetics , Macrolides/pharmacology , Male , Mice , Microtubule-Associated Proteins/deficiency , Microtubule-Associated Proteins/genetics , Mutation , Neurons/drug effects , Progranulins , RNA, Messenger/drug effects , Reverse Transcriptase Polymerase Chain Reaction , Thiazoles/pharmacology
18.
Cells ; 12(1)2022 12 24.
Article in English | MEDLINE | ID: mdl-36611872

ABSTRACT

Amyloid-ß (Aß) deposition is an initiating factor in Alzheimer's disease (AD). Microglia are the brain immune cells that surround and phagocytose Aß plaques, but their phagocytic capacity declines in AD. This is in agreement with studies that associate AD risk loci with genes regulating the phagocytic function of immune cells. Immunotherapies are currently pursued as strategies against AD and there are increased efforts to understand the role of the immune system in ameliorating AD pathology. Here, we evaluated the effect of the Aß targeting ACI-24 vaccine in reducing AD pathology in an amyloidosis mouse model. ACI-24 vaccination elicited a robust and sustained antibody response in APPPS1 mice with an accompanying reduction of Aß plaque load, Aß plaque-associated ApoE and dystrophic neurites as compared to non-vaccinated controls. Furthermore, an increased number of NLRP3-positive plaque-associated microglia was observed following ACI-24 vaccination. In contrast to this local microglial activation at Aß plaques, we observed a more ramified morphology of Aß plaque-distant microglia compared to non-vaccinated controls. Accordingly, bulk transcriptomic analysis revealed a trend towards the reduced expression of several disease-associated microglia (DAM) signatures that is in line with the reduced Aß plaque load triggered by ACI-24 vaccination. Our study demonstrates that administration of the Aß targeting vaccine ACI-24 reduces AD pathology, suggesting its use as a safe and cost-effective AD therapeutic intervention.


Subject(s)
Alzheimer Disease , Amyloidosis , Mice , Animals , Microglia/metabolism , Amyloid beta-Protein Precursor/metabolism , Mice, Transgenic , Amyloid beta-Peptides/metabolism , Alzheimer Disease/genetics , Alzheimer Disease/therapy , Alzheimer Disease/metabolism , Amyloidosis/metabolism , Plaque, Amyloid/metabolism , Phenotype , Vaccination
19.
Cell Rep ; 30(10): 3506-3519.e6, 2020 03 10.
Article in English | MEDLINE | ID: mdl-32160553

ABSTRACT

Genetic variations in TMEM106B, coding for a lysosomal membrane protein, affect frontotemporal lobar degeneration (FTLD) in GRN- (coding for progranulin) and C9orf72-expansion carriers and might play a role in aging. To determine the physiological function of TMEM106B, we generated TMEM106B-deficient mice. These mice develop proximal axonal swellings caused by drastically enlarged LAMP1-positive vacuoles, increased retrograde axonal transport of lysosomes, and accumulation of lipofuscin and autophagosomes. Giant vacuoles specifically accumulate at the distal end and within the axon initial segment, but not in peripheral nerves or at axon terminals, resulting in an impaired facial-nerve-dependent motor performance. These data implicate TMEM106B in mediating the axonal transport of LAMP1-positive organelles in motoneurons and axonal sorting at the initial segment. Our data provide mechanistic insight into how TMEM106B affects lysosomal proteolysis and degradative capacity in neurons.


Subject(s)
Axon Initial Segment/metabolism , Frontotemporal Lobar Degeneration/genetics , Genetic Predisposition to Disease , Lysosomes/metabolism , Membrane Proteins/genetics , Motor Neurons/metabolism , Nerve Tissue Proteins/genetics , Animals , Autophagosomes/metabolism , Autophagosomes/ultrastructure , Axon Initial Segment/ultrastructure , Axonal Transport , Brain Stem/pathology , Cell Nucleus/metabolism , Facial Nerve/pathology , Lysosomes/ultrastructure , Membrane Proteins/deficiency , Mice, Inbred C57BL , Mice, Knockout , Motor Neurons/ultrastructure , Muscles/innervation , Nerve Tissue Proteins/deficiency , Risk Factors
20.
J Neurochem ; 110(3): 1082-94, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19522733

ABSTRACT

Neuronal and glial deposition of misfolded, proteolytically processed, polyubiquitinated and abnormally phosphorylated C-terminal fragments (CTFs) of the TAR DNA binding protein-43 (TDP-43) is a pathological hallmark of frontotemporal lobar degeneration with ubiquitin positive inclusions (FTLD-U) and certain cases of amyotrophic lateral sclerosis. We demonstrate that TDP-43 can be proteolytically processed by caspases upon induction of apoptosis to a major 35 kDa and a minor 25 kDa CTF. These fragments are initially soluble, but over time they accumulate as insoluble and pathologically phosphorylated derivatives. However, proteolytic processing appears not to be absolutely required for the deposition of insoluble TDP-43 species, since a caspase resistant mutant of TDP-43 is also converted into insoluble species. Phosphorylation at S409/410 apparently occurs late during the conversion of soluble to insoluble TDP-43, suggesting that phosphorylation is not a prerequisite for aggregation. Loss of function of the progranulin (PGRN) gene causes FTLD-U with TDP-43 positive inclusions and has been suggested to lead to caspase activation and subsequent TDP-43 processing. However, siRNA-mediated knockdown of PGRN in cell culture as well as a PGRN gene knockout in mice failed to cause the formation of the disease characterizing CTFs of TDP-43. Our findings therefore suggest that caspase-mediated processing generates CTFs of similar biochemical properties as those occurring in nuclear and cytoplasmic deposits of FTLD-U patients independent of PGRN levels.


Subject(s)
Caspases/metabolism , DNA-Binding Proteins/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Animals , Caspases/genetics , Cell Line, Tumor , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , HeLa Cells , Humans , Intercellular Signaling Peptides and Proteins/deficiency , Intercellular Signaling Peptides and Proteins/genetics , Mice , Mice, Knockout , Progranulins
SELECTION OF CITATIONS
SEARCH DETAIL