Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 47
Filter
Add more filters










Publication year range
1.
Front Mol Biosci ; 10: 1169658, 2023.
Article in English | MEDLINE | ID: mdl-37342207

ABSTRACT

Despite the identification of numerous molecular pathways modulating cardiac hypertrophy its pathogenesis is not completely understood. In this study we define an unexpected role for Fibin ("fin bud initiation factor homolog") in cardiomyocyte hypertrophy. Via gene expression profiling in hypertrophic murine hearts after transverse aortic constriction we found a significant induction of Fibin. Moreover, Fibin was upregulated in another mouse model of cardiac hypertrophy (calcineurin-transgenics) as well as in patients with dilated cardiomyopathy. Immunoflourescence microscopy revealed subcellular localization of Fibin at the sarcomeric z-disc. Overexpression of Fibin in neonatal rat ventricular cardiomyocytes revealed a strong anti-hypertrophic effect through inhibiting both, NFAT- and SRF-dependent signalling. In contrast, transgenic mice with cardiac-restricted overexpression of Fibin developed dilated cardiomyopathy, accompanied by induction of hypertrophy-associated genes. Moreover, Fibin overexpression accelerated the progression to heart failure in the presence of prohypertrophic stimuli such as pressure overload and calcineurin overexpression. Histological and ultrastructural analyses surprisingly showed large protein aggregates containing Fibin. On the molecular level, aggregate formation was accompanied by an induction of the unfolded protein response subsequent UPR-mediated apoptosis and autophagy. Taken together, we identified Fibin as a novel potent negative regulator of cardiomyocyte hypertrophy in vitro. Yet, heart-specific Fibin overexpression in vivo causes development of a protein-aggregate-associated cardiomyopathy. Because of close similarities to myofibrillar myopathies, Fibin represents a candidate gene for cardiomyopathy and Fibin transgenic mice may provide additional mechanistic insight into aggregate formation in these diseases.

2.
JACC Basic Transl Sci ; 6(4): 365-380, 2021 Apr.
Article in English | MEDLINE | ID: mdl-33997522

ABSTRACT

Autophagy is a cellular degradation process that has been implicated in diverse disease processes. The authors provide evidence that FYCO1, a component of the autophagic machinery, is essential for adaptation to cardiac stress. Although the absence of FYCO1 does not affect basal autophagy in isolated cardiomyocytes, it abolishes induction of autophagy after glucose deprivation. Likewise, Fyco1-deficient mice subjected to starvation or pressure overload are unable to respond with induction of autophagy and develop impaired cardiac function. FYCO1 overexpression leads to induction of autophagy in isolated cardiomyocytes and transgenic mouse hearts, thereby rescuing cardiac dysfunction in response to biomechanical stress.

3.
J Am Soc Nephrol ; 32(6): 1389-1408, 2021 06 01.
Article in English | MEDLINE | ID: mdl-33785583

ABSTRACT

BACKGROUND: Podocytes embrace the glomerular capillaries with foot processes, which are interconnected by a specialized adherens junction to ultimately form the filtration barrier. Altered adhesion and loss are common features of podocyte injury, which could be mediated by shedding of cell-adhesion molecules through the regulated activity of cell surface-expressed proteases. A Disintegrin and Metalloproteinase 10 (ADAM10) is such a protease known to mediate ectodomain shedding of adhesion molecules, among others. Here we evaluate the involvement of ADAM10 in the process of antibody-induced podocyte injury. METHODS: Membrane proteomics, immunoblotting, high-resolution microscopy, and immunogold electron microscopy were used to analyze human and murine podocyte ADAM10 expression in health and kidney injury. The functionality of ADAM10 ectodomain shedding for podocyte development and injury was analyzed, in vitro and in vivo, in the anti-podocyte nephritis (APN) model in podocyte-specific, ADAM10-deficient mice. RESULTS: ADAM10 is selectively localized at foot processes of murine podocytes and its expression is dispensable for podocyte development. Podocyte ADAM10 expression is induced in the setting of antibody-mediated injury in humans and mice. Podocyte ADAM10 deficiency attenuates the clinical course of APN and preserves the morphologic integrity of podocytes, despite subepithelial immune-deposit formation. Functionally, ADAM10-related ectodomain shedding results in cleavage of the cell-adhesion proteins N- and P-cadherin, thus decreasing their injury-related surface levels. This favors podocyte loss and the activation of downstream signaling events through the Wnt signaling pathway in an ADAM10-dependent manner. CONCLUSIONS: ADAM10-mediated ectodomain shedding of injury-related cadherins drives podocyte injury.


Subject(s)
ADAM10 Protein/metabolism , Amyloid Precursor Protein Secretases/metabolism , Membrane Proteins/metabolism , Nephritis/metabolism , Nephrotic Syndrome/metabolism , Podocytes/metabolism , Podocytes/pathology , Renal Insufficiency, Chronic/metabolism , ADAM10 Protein/genetics , Amyloid Precursor Protein Secretases/genetics , Animals , Autoantibodies/adverse effects , Blood Urea Nitrogen , Cadherins/metabolism , Cell Adhesion , Cell Communication , Cell Membrane/metabolism , Cells, Cultured , Creatinine/urine , Disease Models, Animal , Female , Glomerular Filtration Barrier/pathology , Glomerular Filtration Barrier/physiopathology , Humans , Male , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Nephritis/pathology , Nephrotic Syndrome/pathology , Podocytes/physiology , Proteomics , Tissue Array Analysis , Transcriptome , Wnt Signaling Pathway
4.
Cells ; 9(11)2020 10 31.
Article in English | MEDLINE | ID: mdl-33142804

ABSTRACT

Dysbindin, a schizophrenia susceptibility marker and an essential constituent of BLOC-1 (biogenesis of lysosome-related organelles complex-1), has recently been associated with cardiomyocyte hypertrophy through the activation of Myozap-RhoA-mediated SRF signaling. We employed sandy mice (Dtnbp1_KO), which completely lack Dysbindin protein because of a spontaneous deletion of introns 5-7 of the Dtnbp1 gene, for pathophysiological characterization of the heart. Unlike in vitro, the loss-of-function of Dysbindin did not attenuate cardiac hypertrophy, either in response to transverse aortic constriction stress or upon phenylephrine treatment. Interestingly, however, the levels of hypertrophy-inducing interaction partner Myozap as well as the BLOC-1 partners of Dysbindin like Muted and Pallidin were dramatically reduced in Dtnbp1_KO mouse hearts. Taken together, our data suggest that Dysbindin's role in cardiomyocyte hypertrophy is redundant in vivo, yet essential to maintain the stability of its direct interaction partners like Myozap, Pallidin and Muted.


Subject(s)
Cardiomegaly/genetics , Cardiomegaly/metabolism , Dysbindin/genetics , Dysbindin/metabolism , Muscle Proteins/metabolism , Myocytes, Cardiac/metabolism , Animals , Cytosol/metabolism , Gene Expression Regulation , Hypertrophy/physiopathology , Intracellular Signaling Peptides and Proteins/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Organelle Biogenesis , Protein Binding , Schizophrenia/genetics , Schizophrenia/metabolism , Serum Response Factor/metabolism , Signal Transduction , Vesicular Transport Proteins/metabolism , rhoA GTP-Binding Protein/metabolism
5.
Biochem J ; 477(17): 3433-3451, 2020 09 18.
Article in English | MEDLINE | ID: mdl-32856704

ABSTRACT

Mucopolysaccharidoses comprise a group of rare metabolic diseases, in which the lysosomal degradation of glycosaminoglycans (GAGs) is impaired due to genetically inherited defects of lysosomal enzymes involved in GAG catabolism. The resulting intralysosomal accumulation of GAG-derived metabolites consequently manifests in neurological symptoms and also peripheral abnormalities in various tissues like liver, kidney, spleen and bone. As each GAG consists of differently sulfated disaccharide units, it needs a specific, but also partly overlapping set of lysosomal enzymes to accomplish their complete degradation. Recently, we identified and characterized the lysosomal enzyme arylsulfatase K (Arsk) exhibiting glucuronate-2-sulfatase activity as needed for the degradation of heparan sulfate (HS), chondroitin sulfate (CS) and dermatan sulfate (DS). In the present study, we investigated the physiological relevance of Arsk by means of a constitutive Arsk knockout mouse model. A complete lack of glucuronate desulfation was demonstrated by a specific enzyme activity assay. Arsk-deficient mice show, in an organ-specific manner, a moderate accumulation of HS and CS metabolites characterized by 2-O-sulfated glucuronate moieties at their non-reducing ends. Pathophysiological studies reflect a rather mild phenotype including behavioral changes. Interestingly, no prominent lysosomal storage pathology like bone abnormalities were detected. Our results from the Arsk mouse model suggest a new although mild form of mucopolysacharidose (MPS), which we designate MPS type IIB.


Subject(s)
Arylsulfatases/metabolism , Chondroitin Sulfates/metabolism , Heparitin Sulfate/metabolism , Mucopolysaccharidoses/metabolism , Animals , Arylsulfatases/genetics , Chondroitin Sulfates/genetics , Enzyme Activation , Heparitin Sulfate/genetics , Mice , Mice, Knockout , Mucopolysaccharidoses/genetics
6.
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
7.
Autophagy ; 16(5): 811-825, 2020 05.
Article in English | MEDLINE | ID: mdl-31282275

ABSTRACT

CTSD (cathepsin D) is one of the major lysosomal proteases indispensable for the maintenance of cellular proteostasis by turning over substrates of endocytosis, phagocytosis and autophagy. Consequently, CTSD deficiency leads to a strong impairment of the lysosomal-autophagy machinery. In mice and humans CTSD dysfunction underlies the congenital variant (CLN10) of neuronal ceroid lipofuscinosis (NCL). NCLs are distinct lysosomal storage disorders (LSDs) sharing various hallmarks, namely accumulation of protein aggregates and ceroid lipofuscin leading to neurodegeneration and blindness. The most established and clinically approved approach to treat LSDs is enzyme replacement therapy (ERT) aiming to replace the defective hydrolase with an exogenously applied recombinant protein. Here we reveal that recombinant human pro-CTSD produced in a mammalian expression system can be efficiently taken up by a variety of cell models, is correctly targeted to lysosomes and processed to the active mature form of the protease. In proof-of-principle experiments we provide evidence that recombinant human CTSD (rhCTSD) can improve the biochemical phenotype of CTSD-deficient hippocampal slice cultures in vitro and retinal cells in vivo. Furthermore, we demonstrate that dosing of rhCTSD in the murine CLN10 model leads to a correction of lysosomal hypertrophy, storage accumulation and impaired autophagic flux in the viscera and central nervous system (CNS). We establish that direct delivery of the recombinant protease to the CNS is required for improvement of neuropathology and lifespan extension. Together these data support the continuation of the pre-clinical studies for the application of rhCTSD in the treatment of NCL.Abbreviations: AIF1/IBA1: allograft inflammatory factor 1; BBB: blood brain barrier; CNS: central nervous system; CTSB: cathepsin B; CTSD: cathepsin D; CTSL: cathepsin L; ERT: enzyme replacement therapy; GFAP: glial fibrillary acidic protein; INL: inner nuclear layer; LAMP1: lysosomal-associated membrane protein 1; LAMP2: lysosomal-associated membrane protein 2; MAP1LC3/LC3: microtubule-associated protein 1 light chain 3; LDL: low-density lipoprotein; LRP1: low density lipoprotein receptor-related protein 1; LSD: lysosomal storage disorder; MEFs: mouse embryonic fibroblasts; M6P: mannose 6-phosphate; mCTSD: mature CTSD; NCL: neuronal ceroid lipofuscinosis; ONL: outer nuclear layer; PB: phosphate buffer; proCTSD: pro-cathepsin D; LRPAP1: low density lipoprotein receptor-related protein associated protein 1; rhCTSD: human recombinant CTSD; SAPC: saposin C; SAPD: saposin D; ATP5G1: ATP synthase, H+ transporting, mitochondrial F0 complex, subunit C1 (subunit 9); SQSTM1/p62: sequestosome 1; TPP1: tripeptidyl peptidase I.


Subject(s)
Autophagy/drug effects , Cathepsin D/therapeutic use , Enzyme Replacement Therapy , Neuronal Ceroid-Lipofuscinoses/drug therapy , Neuronal Ceroid-Lipofuscinoses/metabolism , Animals , Brain/drug effects , Brain/metabolism , Cathepsin D/metabolism , Disease Models, Animal , Enzyme Replacement Therapy/methods , Fibroblasts/drug effects , Fibroblasts/metabolism , Humans , Lysosomes/drug effects , Lysosomes/metabolism , Mice, Knockout , Tripeptidyl-Peptidase 1
8.
Elife ; 82019 10 29.
Article in English | MEDLINE | ID: mdl-31661432

ABSTRACT

Lysosomes are major sites for intracellular, acidic hydrolase-mediated proteolysis and cellular degradation. The export of low-molecular-weight catabolic end-products is facilitated by polytopic transmembrane proteins mediating secondary active or passive transport. A number of these lysosomal transporters, however, remain enigmatic. We present a detailed analysis of MFSD1, a hitherto uncharacterized lysosomal family member of the major facilitator superfamily. MFSD1 is not N-glycosylated. It contains a dileucine-based sorting motif needed for its transport to lysosomes. Mfsd1 knockout mice develop splenomegaly and severe liver disease. Proteomics of isolated lysosomes from Mfsd1 knockout mice revealed GLMP as a critical accessory subunit for MFSD1. MFSD1 and GLMP physically interact. GLMP is essential for the maintenance of normal levels of MFSD1 in lysosomes and vice versa. Glmp knockout mice mimic the phenotype of Mfsd1 knockout mice. Our data reveal a tightly linked MFSD1/GLMP lysosomal membrane protein transporter complex.


Subject(s)
Liver/physiology , Lysosomes/metabolism , Membrane Proteins/metabolism , Membrane Transport Proteins/metabolism , Animals , Homeostasis , Membrane Proteins/deficiency , Membrane Proteins/genetics , Membrane Transport Proteins/deficiency , Membrane Transport Proteins/genetics , Mice, Knockout , Protein Binding
9.
Nat Commun ; 10(1): 3521, 2019 08 06.
Article in English | MEDLINE | ID: mdl-31387993

ABSTRACT

The intracellular transport of cholesterol is subject to tight regulation. The structure of the lysosomal integral membrane protein type 2 (LIMP-2, also known as SCARB2) reveals a large cavity that traverses the molecule and resembles the cavity in SR-B1 that mediates lipid transfer. The detection of cholesterol within the LIMP-2 structure and the formation of cholesterol-like inclusions in LIMP-2 knockout mice suggested the possibility that LIMP2 transports cholesterol in lysosomes. We present results of molecular modeling, crosslinking studies, microscale thermophoresis and cell-based assays that support a role of LIMP-2 in cholesterol transport. We show that the cavity in the luminal domain of LIMP-2 can bind and deliver exogenous cholesterol to the lysosomal membrane and later to lipid droplets. Depletion of LIMP-2 alters SREBP-2-mediated cholesterol regulation, as well as LDL-receptor levels. Our data indicate that LIMP-2 operates in parallel with Niemann Pick (NPC)-proteins, mediating a slower mode of lysosomal cholesterol export.


Subject(s)
CD36 Antigens/metabolism , Cholesterol, LDL/metabolism , Lysosomal Membrane Proteins/metabolism , Lysosomes/metabolism , Receptors, Scavenger/metabolism , Animals , CD36 Antigens/genetics , CHO Cells , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cricetulus , Fibroblasts , Gene Knockout Techniques , HeLa Cells , Humans , Intracellular Signaling Peptides and Proteins , Lipid Droplets/metabolism , Lysosomal Membrane Proteins/genetics , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Mice , Niemann-Pick C1 Protein , Protein Domains , RNA, Small Interfering/metabolism , Receptors, Scavenger/genetics
10.
EMBO Rep ; 20(3)2019 03.
Article in English | MEDLINE | ID: mdl-30733280

ABSTRACT

Signal peptide peptidase (SPP) and the four homologous SPP-like (SPPL) proteases constitute a family of intramembrane aspartyl proteases with selectivity for type II-oriented transmembrane segments. Here, we analyse the physiological function of the orphan protease SPPL2c, previously considered to represent a non-expressed pseudogene. We demonstrate proteolytic activity of SPPL2c towards selected tail-anchored proteins. Despite shared ER localisation, SPPL2c and SPP exhibit distinct, though partially overlapping substrate spectra and inhibitory profiles, and are organised in different high molecular weight complexes. Interestingly, SPPL2c is specifically expressed in murine and human testis where it is primarily localised in spermatids. In mice, SPPL2c deficiency leads to a partial loss of elongated spermatids and reduced motility of mature spermatozoa, but preserved fertility. However, matings of male and female SPPL2c-/- mice exhibit reduced litter sizes. Using proteomics we identify the sarco/endoplasmic reticulum Ca2+-ATPase (SERCA2)-regulating protein phospholamban (PLN) as a physiological SPPL2c substrate. Accumulation of PLN correlates with a decrease in intracellular Ca2+ levels in elongated spermatids that likely contribute to the compromised male germ cell differentiation and function of SPPL2c-/- mice.


Subject(s)
Aspartic Acid Endopeptidases/metabolism , Calcium-Binding Proteins/metabolism , Cell Membrane/enzymology , Germ Cells/metabolism , Membrane Proteins/metabolism , Amino Acid Sequence , Animals , Aspartic Acid Endopeptidases/chemistry , Calcium/metabolism , Endoplasmic Reticulum/metabolism , Female , HEK293 Cells , HeLa Cells , Homeostasis , Humans , Male , Membrane Proteins/chemistry , Mice , Organ Specificity , Spermatids/metabolism , Substrate Specificity , Testis/enzymology
11.
Oncotarget ; 8(27): 43635-43652, 2017 Jul 04.
Article in English | MEDLINE | ID: mdl-28504966

ABSTRACT

The Transmembrane protein 192 (TMEM192) is a lysosomal/late endosomal protein initially discovered by organellar proteomics. TMEM192 exhibits four transmembrane segments with cytosolic N- and C-termini and forms homodimers. Devoid of significant homologies, the molecular function of TMEM192 is currently unknown. Upon TMEM192 knockdown in hepatoma cells, a dysregulation of autophagy and increased apoptosis were reported. Here, we aimed to define the physiological role of TMEM192 by analysing consequences of TMEM192 ablation in mice. Therefore, we compared the biochemical properties of murine TMEM192 to those of the human orthologue. We reveal lysosomal residence of murine TMEM192 and demonstrate ubiquitous tissue expression. In brain, TMEM192 expression was pronounced in the hippocampus but also present in the cortex and cerebellum, as analysed based on a lacZ reporter allele. Murine TMEM192 undergoes proteolytic processing in a tissue-specific manner. Thereby, a 17 kDa fragment is generated which was detected in most murine tissues except liver. TMEM192 processing occurs after lysosomal targeting by pH-dependent lysosomal proteases. TMEM192-/- murine embryonic fibroblasts (MEFs) exhibited a regular morphology of endo-/lysosomes and were capable of performing autophagy and lysosomal exocytosis. Histopathological, ultrastructural and biochemical analyses of all major tissues of TMEM192-/- mice demonstrated normal lysosomal functions without apparent lysosomal storage. Furthermore, the abundance of the major immune cells was comparable in TMEM192-/- and wild type mice. Based on this, we conclude that under basal conditions in vivo the loss of TMEM192 can be efficiently compensated by alternative pathways. Further studies will be required to decipher its molecular function.


Subject(s)
Intracellular Membranes/metabolism , Lysosomes/metabolism , Membrane Proteins/metabolism , Animals , Autophagy , Brain/metabolism , Brain/pathology , Cell Line, Tumor , Fibroblasts , Gene Expression , Gene Knockout Techniques , Glycosylation , Liver/metabolism , Liver/pathology , Membrane Proteins/genetics , Mice , Mice, Knockout , Proteolysis , Spleen/metabolism , Spleen/pathology , Ubiquitination
12.
J Immunol ; 199(1): 172-185, 2017 07 01.
Article in English | MEDLINE | ID: mdl-28550201

ABSTRACT

The invariant chain (CD74) mediates assembly and targeting of MHC class II (MHCII) complexes. In endosomes, CD74 undergoes sequential degradation by different proteases, including cathepsin S (CatS) and the intramembrane protease signal peptide peptidase-like 2a (SPPL2a). In their absence, CD74 N-terminal fragments (NTFs) accumulate. In SPPL2a-/- B cells, such an NTF impairs endosomal trafficking and BCR signal transduction. In mice, this leads to a loss of splenic B cells beyond the transitional stage 1. To gain insight into CD74 determinants and the role of MHCII, we compared B cells from CatS-/- , SPPL2a-/- , and SPPL2a-MHCII double-deficient mice. We assessed differentiation of B cells in bone marrow and spleen and analyzed their endosomal morphology, BCR expression, and signal transduction. We demonstrate that MHCII is dispensable for the B cell phenotype of SPPL2a-/- mice, further supporting a CD74-intrinsic effect. Despite significant vacuolization of endosomal compartments similar to SPPL2a-/- B cells, CatS-/- traditional stage 1 B cells show unimpaired degradation of endocytic cargo, have intact BCR signaling, and do not exhibit any relevant defects in maturation. This could indicate that CD74 NTF-induced structural changes of endosomes are not directly involved in these processes. We further found that the block of CD74 degradation in CatS-/- B cells is incomplete, so that NTF levels are significantly lower than in SPPL2a-/- B cells. This suggests a dose dependency and threshold for the CD74 NTF-associated impairment of B cell signaling and maturation. In addition, different functional properties of the longer, MHCII-bound CD74 NTF could contribute to the milder phenotype of CatS-/- B cells.


Subject(s)
Antigens, Differentiation, B-Lymphocyte/immunology , B-Lymphocytes/immunology , Genes, MHC Class II , Histocompatibility Antigens Class II/immunology , Animals , Antigens, Differentiation, B-Lymphocyte/metabolism , Aspartic Acid Endopeptidases/deficiency , Aspartic Acid Endopeptidases/genetics , Aspartic Acid Endopeptidases/metabolism , Cathepsins/deficiency , Cathepsins/genetics , Cathepsins/metabolism , Cell Differentiation , Endosomes/immunology , Endosomes/physiology , Histocompatibility Antigens Class II/metabolism , Membrane Proteins/deficiency , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Signal Transduction
13.
Autophagy ; 13(4): 670-685, 2017 Apr 03.
Article in English | MEDLINE | ID: mdl-28129027

ABSTRACT

The vacuolar-type H+-translocating ATPase (v-H+-ATPase) has been implicated in the amino acid-dependent activation of the mechanistic target of rapamycin complex 1 (MTORC1), an important regulator of macroautophagy. To reveal the mechanistic links between the v-H+-ATPase and MTORC1, we destablilized v-H+-ATPase complexes in mouse liver cells by induced deletion of the essential chaperone ATP6AP2. ATP6AP2-mutants are characterized by massive accumulation of endocytic and autophagic vacuoles in hepatocytes. This cellular phenotype was not caused by a block in endocytic maturation or an impaired acidification. However, the degradation of LC3-II in the knockout hepatocytes appeared to be reduced. When v-H+-ATPase levels were decreased, we observed lysosome association of MTOR and normal signaling of MTORC1 despite an increase in autophagic marker proteins. To better understand why MTORC1 can be active when v-H+-ATPase is depleted, the activation of MTORC1 was analyzed in ATP6AP2-deficient fibroblasts. In these cells, very little amino acid-elicited activation of MTORC1 was observed. In contrast, insulin did induce MTORC1 activation, which still required intracellular amino acid stores. These results suggest that in vivo the regulation of macroautophagy depends not only on v-H+-ATPase-mediated regulation of MTORC1.


Subject(s)
Autophagy , Liver/enzymology , Lysosomes/metabolism , Mechanistic Target of Rapamycin Complex 1/metabolism , Proton-Translocating ATPases/metabolism , Receptors, Cell Surface/metabolism , Vacuoles/enzymology , Amino Acids/pharmacology , Animals , Autophagy/drug effects , Cells, Cultured , Embryo, Mammalian/cytology , Endocytosis/drug effects , Endosomes/drug effects , Endosomes/metabolism , Fibroblasts/drug effects , Fibroblasts/enzymology , Hepatocytes/drug effects , Hepatocytes/enzymology , Insulin/pharmacology , Liver/drug effects , Liver/ultrastructure , Lysosomes/drug effects , Mice, Knockout , Proton-Translocating ATPases/deficiency , Receptors, Cell Surface/deficiency , Vacuoles/drug effects
14.
Dis Model Mech ; 9(9): 1015-28, 2016 09 01.
Article in English | MEDLINE | ID: mdl-27491075

ABSTRACT

Fucosidosis is a rare lysosomal storage disorder caused by the inherited deficiency of the lysosomal hydrolase α-L-fucosidase, which leads to an impaired degradation of fucosylated glycoconjugates. Here, we report the generation of a fucosidosis mouse model, in which the gene for lysosomal α-L-fucosidase (Fuca1) was disrupted by gene targeting. Homozygous knockout mice completely lack α-L-fucosidase activity in all tested organs leading to highly elevated amounts of the core-fucosylated glycoasparagine Fuc(α1,6)-GlcNAc(ß1-N)-Asn and, to a lesser extent, other fucosylated glycoasparagines, which all were also partially excreted in urine. Lysosomal storage pathology was observed in many visceral organs, such as in the liver, kidney, spleen and bladder, as well as in the central nervous system (CNS). On the cellular level, storage was characterized by membrane-limited cytoplasmic vacuoles primarily containing water-soluble storage material. In the CNS, cellular alterations included enlargement of the lysosomal compartment in various cell types, accumulation of secondary storage material and neuroinflammation, as well as a progressive loss of Purkinje cells combined with astrogliosis leading to psychomotor and memory deficits. Our results demonstrate that this new fucosidosis mouse model resembles the human disease and thus will help to unravel underlying pathological processes. Moreover, this model could be utilized to establish diagnostic and therapeutic strategies for fucosidosis.


Subject(s)
Brain/pathology , Fucosidosis/metabolism , Fucosidosis/pathology , Animals , Behavior, Animal , Brain/ultrastructure , Disease Models, Animal , Enzyme Activation , Fucose/metabolism , Fucosidosis/urine , G(M2) Ganglioside/metabolism , Glycoconjugates/urine , Glycoproteins/metabolism , Humans , Inflammation/pathology , Lysosomes/enzymology , Lysosomes/ultrastructure , Mice, Inbred C57BL , Organ Specificity , Proteolysis , Purkinje Cells/metabolism , Purkinje Cells/pathology , Viscera/metabolism , Viscera/pathology , alpha-L-Fucosidase/deficiency , alpha-L-Fucosidase/metabolism
15.
Cardiovasc Res ; 110(3): 381-94, 2016 06 01.
Article in English | MEDLINE | ID: mdl-27056896

ABSTRACT

AIMS: Down syndrome-associated dual-specificity tyrosine-(Y)-phosphorylation-regulated kinase 1A (DYRK1A) is a ubiquitously expressed protein kinase. Up to date a variety of targets have been identified, establishing a key role for Dyrk1a in selected signalling pathways. In cardiomyocytes, Dyrk1a acts as a negative regulator of hypertrophy by phosphorylating transcription factors of the NFAT family, but its mechanistic function in the heart remains poorly understood. This study was designed to investigate a potential protective role of Dyrk1a in cardiac hypertrophy in vivo. METHODS AND RESULTS: We generated transgenic mice with cardiac-specific overexpression of Dyrk1a. Counterintuitively, these mice developed severe dilated cardiomyopathy associated with congestive heart failure and premature death. In search for the cause of this unexpected phenotype, we found that Dyrk1a interacts with all members of the D-cyclin family and represses their protein levels in vitro and in vivo. Particularly, forced expression of Dyrk1a leads to increased phosphorylation of Ccnd2 on Thr280 and promotes its subsequent proteasomal degradation. Accordingly, cardiomyocytes overexpressing Dyrk1a display hypo-phosphorylated Rb1, suppression of Rb/E2f-signalling, and reduced expression of E2f-target genes, which ultimately results in impaired cell cycle progression. CONCLUSIONS: We identified Dyrk1a as a novel negative regulator of D-cyclin-mediated Rb/E2f-signalling. As dysregulation of this pathway with impaired cardiomyocyte proliferation leads to cardiomyopathy, dose-specific Dyrk1a expression and activity appears to be critical for the hyperplastic and hypertrophic growth of the developing heart.


Subject(s)
Cardiomegaly/enzymology , Cardiomyopathy, Dilated/enzymology , Cyclin D/metabolism , E2F Transcription Factors/metabolism , Myocytes, Cardiac/enzymology , Protein Serine-Threonine Kinases/metabolism , Protein-Tyrosine Kinases/metabolism , Retinoblastoma/metabolism , Signal Transduction , Animals , Cardiomegaly/genetics , Cardiomegaly/pathology , Cardiomegaly/physiopathology , Cardiomyopathy, Dilated/genetics , Cardiomyopathy, Dilated/pathology , Cardiomyopathy, Dilated/physiopathology , Cell Cycle , Cell Proliferation , Cyclin D/genetics , Disease Models, Animal , Gene Expression Regulation , HEK293 Cells , Heart Failure/enzymology , Heart Failure/genetics , Heart Failure/pathology , Heart Failure/physiopathology , Humans , Mice, Inbred C57BL , Mice, Transgenic , Myocytes, Cardiac/pathology , Phosphorylation , Protein Binding , Protein Serine-Threonine Kinases/genetics , Protein-Tyrosine Kinases/genetics , Rats, Wistar , Time Factors , Transfection , Dyrk Kinases
16.
Invest Ophthalmol Vis Sci ; 57(3): 1120-31, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26975023

ABSTRACT

PURPOSE: Retinal degeneration is a common feature of several lysosomal storage disorders, including the mucopolysaccharidoses, a group of metabolic disorders that is characterized by widespread accumulation of glycosaminoglycans due to lysosomal enzyme dysfunction. We used a new mouse model of mucopolysaccharidosis IIIE to study the effect of Arylsulfatase G (ARSG) deficiency on retina integrity. METHODS: The retina of Arsg knockout mice aged 1 to 24 months was studied by immunohistochemistry and Western blot analysis. Electron microscopic analyses were performed on retinas from 15- and 22-month-old animals. Photoreceptor and microglia cell numbers and retina thickness were determined to quantitatively characterize retinal degeneration in ARSG-deficient mice. RESULTS: Arsg knockout mice showed a progressive degeneration of photoreceptor cells starting between 1 and 6 months of age, resulting in the loss of more than 50% of photoreceptor cells in 24-month-old mice. Photoreceptor loss was accompanied by reactive astrogliosis, reactive microgliosis that was evident in the outer but not inner retina, and elevated expression levels of some lysosomal proteins. Electron microscopic analyses of retinas revealed no evidence for the presence of storage vacuoles. Of note, expression of ARSG protein in wild-type mice was detectable only in the RPE which, however, appeared morphologically unaffected in knockout mice at the electron microscopic level. CONCLUSIONS: To our knowledge, this is the first study demonstrating that ARSG deficiency results in progressive photoreceptor degeneration and dysregulation of various lysosomal proteins.


Subject(s)
Arylsulfatases/deficiency , Disease Models, Animal , Mucopolysaccharidosis III/enzymology , Photoreceptor Cells/enzymology , Retinal Degeneration/enzymology , Animals , Arylsulfatases/metabolism , Blotting, Western , Immunohistochemistry , Mice , Mice, Inbred C57BL , Mice, Knockout , Microglia/pathology , Mucopolysaccharidosis III/diagnosis , Photoreceptor Cells/pathology , Proteins/metabolism , Retinal Degeneration/diagnosis , beta-N-Acetylhexosaminidases/metabolism
17.
Oncotarget ; 7(14): 17431-41, 2016 Apr 05.
Article in English | MEDLINE | ID: mdl-26942887

ABSTRACT

UNLABELLED: A Disintegrin And Metalloprotease (ADAM) 10 exerts essential roles during organ development and tissue integrity in different organs, mainly through activation of the Notch pathway. However, only little is known about its implication in liver tissue physiology. Here we show that in contrast to its role in other tissues, ADAM10 is dispensable for the Notch2-dependent biliary tree formation. However, we demonstrate that expression of bile acid transporters is dependent on ADAM10. Consequently, mice deficient for Adam10 in hepatocytes, cholangiocytes and liver progenitor cells develop spontaneous hepatocyte necrosis and concomitant liver fibrosis. We furthermore observed a strongly augmented ductular reaction in 15-week old ADAM10(Δhep/Δch) mice and demonstrate that c-Met dependent liver progenitor cell activation is enhanced. Additionally, liver progenitor cells are primed to hepatocyte differentiation in the absence of ADAM10. These findings show that ADAM10 is a novel central node controlling liver tissue homeostasis. HIGHLIGHTS: Loss of ADAM10 in murine liver results in hepatocyte necrosis and concomitant liver fibrosis. ADAM10 directly regulates expression of bile acid transporters but is dispensable for Notch2-dependent formation of the biliary system. Activation of liver progenitor cells is enhanced through increased c-Met signalling, in the absence of ADAM10. Differentiation of liver progenitor cells to hepatocytes is augmented in the absence of ADAM10.


Subject(s)
ADAM10 Protein/metabolism , Amyloid Precursor Protein Secretases/metabolism , Liver/metabolism , Membrane Proteins/metabolism , ADAM10 Protein/deficiency , ADAM10 Protein/genetics , Amyloid Precursor Protein Secretases/deficiency , Amyloid Precursor Protein Secretases/genetics , Animals , Carrier Proteins/metabolism , Cell Differentiation/physiology , Cell Proliferation/physiology , Down-Regulation , Hepatocytes/metabolism , Hepatocytes/pathology , Homeostasis , Liver/cytology , Liver/pathology , Membrane Glycoproteins/metabolism , Membrane Proteins/deficiency , Membrane Proteins/genetics , Mice , Mice, Knockout , Mice, Transgenic , Necrosis , Receptor, Notch2/metabolism , Signal Transduction
18.
J Biol Chem ; 291(8): 4128-43, 2016 Feb 19.
Article in English | MEDLINE | ID: mdl-26719331

ABSTRACT

The intercalated disc (ID) is a "hot spot" for heart disease, as several ID proteins have been found mutated in cardiomyopathy. Myozap is a recent addition to the list of ID proteins and has been implicated in serum-response factor signaling. To elucidate the cardiac consequences of targeted deletion of myozap in vivo, we generated myozap-null mutant (Mzp(-/-)) mice. Although Mzp(-/-) mice did not exhibit a baseline phenotype, increased biomechanical stress due to pressure overload led to accelerated cardiac hypertrophy, accompanied by "super"-induction of fetal genes, including natriuretic peptides A and B (Nppa/Nppb). Moreover, Mzp(-/-) mice manifested a severe reduction of contractile function, signs of heart failure, and increased mortality. Expression of other ID proteins like N-cadherin, desmoplakin, connexin-43, and ZO-1 was significantly perturbed upon pressure overload, underscored by disorganization of the IDs in Mzp(-/-) mice. Exploration of the molecular causes of enhanced cardiac hypertrophy revealed significant activation of ß-catenin/GSK-3ß signaling, whereas MAPK and MKL1/serum-response factor pathways were inhibited. In summary, myozap is required for proper adaptation to increased biomechanical stress. In broader terms, our data imply an essential function of the ID in cardiac remodeling beyond a mere structural role and emphasize the need for a better understanding of this molecular structure in the context of heart disease.


Subject(s)
Cardiomegaly/metabolism , Glycogen Synthase Kinase 3/metabolism , MAP Kinase Signaling System , Muscle Proteins/metabolism , Serum Response Factor/metabolism , Trans-Activators/metabolism , beta Catenin/metabolism , Animals , Cardiomegaly/genetics , Cardiomegaly/pathology , Extracellular Signal-Regulated MAP Kinases/genetics , Extracellular Signal-Regulated MAP Kinases/metabolism , Glycogen Synthase Kinase 3/genetics , Glycogen Synthase Kinase 3 beta , Mice , Mice, Knockout , Muscle Proteins/genetics , Rats , Serum Response Factor/genetics , Trans-Activators/genetics , Transcription Factors , beta Catenin/genetics
19.
Cell Mol Gastroenterol Hepatol ; 1(6): 678-694, 2015 Nov 01.
Article in English | MEDLINE | ID: mdl-26693174

ABSTRACT

BACKGROUND & AIMS: The pathogenic mechanism of pancreatitis is poorly understood. Recent evidence implicates defective autophagy in pancreatitis responses; however, the pathways mediating impaired autophagy in pancreas remain largely unknown. Here, we investigate the role of lysosome associated membrane proteins (LAMPs) in pancreatitis. METHODS: We analyzed changes in LAMPs in experimental models and human pancreatitis, and the underlying mechanisms: LAMP de-glycosylation and degradation. LAMP cleavage by cathepsin B (CatB) was analyzed by mass spectrometry. We used mice deficient in LAMP-2 to assess its role in pancreatitis. RESULTS: Pancreatic levels of LAMP-1 and LAMP-2 greatly decrease across various pancreatitis models and in human disease. Pancreatitis does not trigger LAMPs' bulk de-glycosylation, but induces their degradation via CatB-mediated cleavage of LAMP molecule close to the boundary between luminal and transmembrane domains. LAMP-2 null mice spontaneously develop pancreatitis that begins with acinar cell vacuolization due to impaired autophagic flux, and progresses to severe pancreas damage characterized by trypsinogen activation, macrophage-driven inflammation, and acinar cell death. LAMP-2 deficiency causes a decrease in pancreatic digestive enzymes content, stimulates the basal and inhibits CCK-induced amylase secretion by acinar cells. The effects of LAMP-2 knockout and acute cerulein pancreatitis overlap, which corroborates the pathogenic role of LAMP decrease in experimental pancreatitis models. CONCLUSIONS: The results indicate a critical role for LAMPs, particularly LAMP-2, in maintaining pancreatic acinar cell homeostasis, and provide evidence that defective lysosomal function, resulting in impaired autophagy, leads to pancreatitis. Mice with LAMP-2 deficiency present a novel genetic model of human pancreatitis caused by lysosomal/autophagic dysfunction.

20.
Hum Mol Genet ; 24(7): 1856-68, 2015 Apr 01.
Article in English | MEDLINE | ID: mdl-25452429

ABSTRACT

Deficiency of arylsulfatase G (ARSG) leads to a lysosomal storage disease in mice resembling biochemical and pathological features of the mucopolysaccharidoses and particularly features of mucopolysaccharidosis type III (Sanfilippo syndrome). Here we show that Arsg KO mice share common neuropathological findings with other Sanfilippo syndrome models and patients, but they can be clearly distinguished by the limitation of most phenotypic alterations to the cerebellum, presenting with ataxia as the major neurological finding. We determined in detail the expression of ARSG in the central nervous system and observed highest expression in perivascular macrophages (which are characterized by abundant vacuolization in Arsg KO mice) and oligodendrocytes. To gain insight into possible mechanisms leading to ataxia, the pathology in older adult mice (>12 months) was investigated in detail. This study revealed massive loss of Purkinje cells and gliosis in the cerebellum, and secondary accumulation of glycolipids like GM2 and GM3 gangliosides and unesterified cholesterol in surviving Purkinje cells, as well as neurons of some other brain regions. The abundant presence of ubiquitin and p62-positive aggregates in degenerating Purkinje cells coupled with the absence of significant defects in macroautophagy is consistent with lysosomal membrane permeabilization playing a role in the pathogenesis of Arsg-deficient mice and presumably Sanfilippo disease in general. Our data delineating the phenotype of mucopolysaccharidosis IIIE in a mouse KO model should help in the identification of possible human cases of this disease.


Subject(s)
Arylsulfatases/deficiency , Ataxia/enzymology , Mucopolysaccharidosis III/enzymology , Animals , Arylsulfatases/genetics , Ataxia/genetics , Ataxia/metabolism , Ataxia/pathology , Cerebellum/cytology , Cerebellum/metabolism , Disease Models, Animal , Female , Gliosis/metabolism , Glycolipids/metabolism , Humans , Male , Mice , Mice, Knockout , Mucopolysaccharidosis III/genetics , Mucopolysaccharidosis III/metabolism , Mucopolysaccharidosis III/pathology , Purkinje Cells/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...