Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
1.
Contact (Thousand Oaks) ; 7: 25152564241244941, 2024.
Article in English | MEDLINE | ID: mdl-38585502

ABSTRACT

Changes in the connections between the endoplasmic reticulum (ER) and mitochondria, as well as alterations in mitochondria-associated ER membrane (MAM) signalling, have been documented in various neurodegenerative diseases affecting the brain. Despite the growing recognition of the significance of the gut-brain axis in neurodegenerative conditions, there has been no prior investigation into the biology of MAM within the enteric nervous system (ENS). Our recent research reveals, for the first time, the existence of connections between the ER and mitochondria within enteric neurons. Additionally, we observed alterations in the dynamics of these connections in the enteric neurons from a mouse model exhibiting age-related neurodegeneration. These findings provide the first detailed characterization of MAM in the ENS under physiological conditions and in a mouse model of age-associated neurodegeneration and shed new light on the potential role of enteric MAM in the context of neurodegenerative disorders.

2.
Acta Neuropathol Commun ; 12(1): 32, 2024 Feb 23.
Article in English | MEDLINE | ID: mdl-38395965

ABSTRACT

Frontotemporal dementia (FTD) and amyotrophic lateral sclerosis (ALS) are clinically linked major neurodegenerative diseases. Notably, TAR DNA-binding protein-43 (TDP43) accumulations are hallmark pathologies of FTD/ALS and mutations in the gene encoding TDP43 cause familial FTD/ALS. There are no cures for FTD/ALS. FTD/ALS display damage to a broad range of physiological functions, many of which are regulated by signaling between the endoplasmic reticulum (ER) and mitochondria. This signaling is mediated by the VAPB-PTPIP51 tethering proteins that serve to recruit regions of ER to the mitochondrial surface so as to facilitate inter-organelle communications. Several studies have now shown that disrupted ER-mitochondria signaling including breaking of the VAPB-PTPIP51 tethers are features of FTD/ALS and that for TDP43 and other familial genetic FTD/ALS insults, this involves activation of glycogen kinase-3ß (GSK3ß). Such findings have prompted suggestions that correcting damage to ER-mitochondria signaling and the VAPB-PTPIP51 interaction may be broadly therapeutic. Here we provide evidence to support this notion. We show that overexpression of VAPB or PTPIP51 to enhance ER-mitochondria signaling corrects mutant TDP43 induced damage to inositol 1,4,5-trisphosphate (IP3) receptor delivery of Ca2+ to mitochondria which is a primary function of the VAPB-PTPIP51 tethers, and to synaptic function. Moreover, we show that ursodeoxycholic acid (UDCA), an FDA approved drug linked to FTD/ALS and other neurodegenerative diseases therapy and whose precise therapeutic target is unclear, corrects TDP43 linked damage to the VAPB-PTPIP51 interaction. We also show that this effect involves inhibition of TDP43 mediated activation of GSK3ß. Thus, correcting damage to the VAPB-PTPIP51 tethers may have therapeutic value for FTD/ALS and other age-related neurodegenerative diseases.


Subject(s)
Amyotrophic Lateral Sclerosis , Frontotemporal Dementia , Neurodegenerative Diseases , Vesicular Transport Proteins , Humans , Amyotrophic Lateral Sclerosis/pathology , Calcium/metabolism , Endoplasmic Reticulum/metabolism , Frontotemporal Dementia/genetics , Frontotemporal Dementia/metabolism , Glycogen Synthase Kinase 3 beta/metabolism , Mitochondria/metabolism , Neurodegenerative Diseases/metabolism , Protein Tyrosine Phosphatases/metabolism , Synapses/pathology , TDP-43 Proteinopathies/metabolism , Vesicular Transport Proteins/genetics
3.
Am J Physiol Gastrointest Liver Physiol ; 326(3): G330-G343, 2024 Mar 01.
Article in English | MEDLINE | ID: mdl-38226933

ABSTRACT

Alterations in endoplasmic reticulum (ER)-mitochondria associations and in mitochondria-associated ER membrane (MAM) behavior have been reported in the brain in several neurodegenerative diseases. Despite the emerging role of the gut-brain axis in neurodegenerative disorders, the biology of MAM in the enteric nervous system (ENS) has not previously been studied. Therefore, we set out to characterize the MAM in the distal colon of wild-type C57BL/6J mice and senescence-accelerated mouse prone 8 (SAMP8), a mouse model of age-related neurodegeneration. We showed for the first time that MAMs are widely present in enteric neurons and that their association is altered in SAMP8 mice. We then examined the functions of MAMs in a primary culture model of enteric neurons and showed that calcium homeostasis was altered in SAMP8 mice when compared with control animals. These findings provide the first detailed characterization of MAMs in the ENS under physiological conditions and during age-associated neurodegeneration. Further investigation of MAM modifications in the ENS in disease may provide valuable information about the possible role of enteric MAMs in neurodegenerative diseases.NEW & NOTEWORTHY Our work shows for the first time the presence of contacts between endoplasmic reticulum and mitochondria in the enteric neurons and that the dynamic of these contacts is affected in these cells from an age-related neurodegeneration mouse model. It provides new insights into the potential role of enteric mitochondria-associated endoplasmic reticulum membrane in neurodegenerative disorders.


Subject(s)
Enteric Nervous System , Neurodegenerative Diseases , Mice , Animals , Mitochondria Associated Membranes , Mice, Inbred C57BL , Endoplasmic Reticulum , Disease Models, Animal
4.
Front Cell Dev Biol ; 10: 920947, 2022.
Article in English | MEDLINE | ID: mdl-36120587

ABSTRACT

Signaling between the endoplasmic reticulum (ER) and mitochondria regulates a number of fundamental physiological processes. This signaling involves close physical contacts between the two organelles that are mediated by the VAPB-PTPIP51 ″tethering" proteins. The VAPB-PTPIP51 tethers facilitate inositol 1,4,5-trisphosphate (IP3) receptor delivery of Ca2+ from ER to mitochondria. Damage to the tethers is seen in Alzheimer's disease, Parkinson's disease and frontotemporal dementia with related amyotrophic lateral sclerosis (FTD/ALS). Understanding the mechanisms that regulate the VAPB-PTPIP51 interaction thus represents an important area of research. Recent studies suggest that an FFAT motif in PTPIP51 is key to its binding to VAPB but this work relies on in vitro studies with short peptides. Cellular studies to support this notion with full-length proteins are lacking. Here we address this issue. Immunoprecipitation assays from transfected cells revealed that deletion of the PTPIP51 FFAT motif has little effect on VAPB binding. However, mutation and deletion of a nearby coiled-coil domain markedly affect this binding. Using electron microscopy, we then show that deletion of the coiled-coil domain but not the FFAT motif abrogates the effect of PTPIP51 on ER-mitochondria contacts. Finally, we show that deletion of the coiled-coil domain but not the FFAT motif abrogates the effect of PTPIP51 on the IP3 receptor-mediated delivery of Ca2+ to mitochondria. Thus, the coiled-coil domain is essential for PTPIP51 ER-mitochondria signaling functions.

5.
Neurogastroenterol Motil ; 34(6): e14354, 2022 06.
Article in English | MEDLINE | ID: mdl-35279896

ABSTRACT

BACKGROUND: It is now well established that phosphorylated alpha-synuclein histopathology, the pathologic hallmark of Parkinson's disease (PD) is not limited to the brain but also extends to the enteric nervous system (ENS). This observation led to the hypothesis that the ENS could play a pivotal role in the development of PD. Research on the enteric synucleinopathy has, however, been hampered by difficulties in detecting phosphorylated alpha-synuclein in the ENS by Western blotting, even when the transferred membrane is fixed with an optimized protocol. This suggests that the available antibodies used in previous studies lacked of sensitivity for the detection of phosphorylated alpha-synuclein at Ser129 in enteric neurons. Here, we evaluated three recent commercially available phospho-alpha-synuclein antibodies and compared them to two antibodies used in previous research. METHODS: The specificity and sensitivity of the 5 antibodies were evaluated by Western blot performed with recombinant alpha-synuclein and with protein lysates from rat primary cultures of ENS. In primary culture of ENS, additional experiments were performed with the most specific antibody in order to modulate alpha-synuclein phosphorylation and to validate its utilization in immunofluorescence experiments. RESULTS: The rabbit monoclonal antibody D1R1R uniquely and robustly detected endogenous phosphorylated alpha-synuclein at Ser129 in rat primary culture of ENS without any non-specific bands, allowing for a reliable analysis of phosphorylated alpha-synuclein regulation by pharmacologic means. CONCLUSIONS AND INFERENCES: Using D1R1R antibody together with the optimized protocol for membrane fixation may help deciphering the signaling pathways involved in enteric alpha-synuclein post-translational regulation in PD.


Subject(s)
Enteric Nervous System , Parkinson Disease , Animals , Blotting, Western , Enteric Nervous System/metabolism , Neurons/metabolism , Parkinson Disease/metabolism , Rats , alpha-Synuclein
6.
J Cell Sci ; 135(3)2022 02 01.
Article in English | MEDLINE | ID: mdl-35129196

ABSTRACT

Recent advances have revealed common pathological changes in neurodegenerative diseases, such as Alzheimer's disease, Parkinson's disease and amyotrophic lateral sclerosis with related frontotemporal dementia (ALS/FTD). Many of these changes can be linked to alterations in endoplasmic reticulum (ER)-mitochondria signaling, including dysregulation of Ca2+ signaling, autophagy, lipid metabolism, ATP production, axonal transport, ER stress responses and synaptic dysfunction. ER-mitochondria signaling involves specialized regions of ER, called mitochondria-associated membranes (MAMs). Owing to their role in neurodegenerative processes, MAMs have gained attention as they appear to be associated with all the major neurodegenerative diseases. Furthermore, their specific role within neuronal maintenance is being revealed as mutant genes linked to major neurodegenerative diseases have been associated with damage to these specialized contacts. Several studies have now demonstrated that these specialized contacts regulate neuronal health and synaptic transmission, and that MAMs are damaged in patients with neurodegenerative diseases. This Review will focus on the role of MAMs and ER-mitochondria signaling within neurons and how damage of the ER-mitochondria axis leads to a disruption of vital processes causing eventual neurodegeneration.


Subject(s)
Amyotrophic Lateral Sclerosis , Frontotemporal Dementia , Neurodegenerative Diseases , Amyotrophic Lateral Sclerosis/metabolism , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum Stress , Frontotemporal Dementia/metabolism , Humans , Mitochondria/metabolism , Neurodegenerative Diseases/metabolism , Neurons/metabolism
7.
J Neural Transm (Vienna) ; 129(9): 1095-1103, 2022 09.
Article in English | MEDLINE | ID: mdl-34816335

ABSTRACT

Alpha-synuclein deposits, the pathological hallmarks of Parkinson's disease, are consistently found in the gastrointestinal tract of parkinsonian subjects. These observations have raised the potential that endoscopically obtainable mucosal biopsies can aid to a molecular diagnosis of the disease. The possible usefulness of mucosal biopsies is, however, not limited to the detection of alpha-synuclein, but also extends to other essential aspects underlying pathophysiological mechanisms of gastrointestinal manifestations in Parkinson's disease. The aim of the current review is to provide an appraisal of the existing studies showing that gastrointestinal biopsies can be used for the analysis of enteric neuronal and glial cell morphology, intestinal epithelial barrier function, and gastrointestinal inflammation in Parkinson's disease. A perspective on the generation of organoids with GI biopsies and the potential use of single-cell and spatial transcriptomic technologies will be also addressed.


Subject(s)
Parkinson Disease , alpha-Synuclein , Biopsy , Gastrointestinal Tract/chemistry , Gastrointestinal Tract/pathology , Humans , Neurons/pathology , Parkinson Disease/diagnosis , alpha-Synuclein/analysis
8.
Neurobiol Dis ; 143: 105020, 2020 09.
Article in English | MEDLINE | ID: mdl-32682953

ABSTRACT

Signaling between the endoplasmic reticulum (ER) and mitochondria regulates a number of key neuronal functions, many of which are perturbed in Alzheimer's disease. Moreover, damage to ER-mitochondria signaling is seen in cell and transgenic models of Alzheimer's disease. However, as yet there is little evidence that ER-mitochondria signaling is altered in human Alzheimer's disease brains. ER-mitochondria signaling is mediated by interactions between the integral ER protein VAPB and the outer mitochondrial membrane protein PTPIP51 which act to recruit and "tether" regions of ER to the mitochondrial surface. The VAPB-PTPIP51 tethers are now known to regulate a number of ER-mitochondria signaling functions including delivery of Ca2+from ER stores to mitochondria, mitochondrial ATP production, autophagy and synaptic activity. Here we investigate the VAPB-PTPIP51 tethers in post-mortem control and Alzheimer's disease brains. Quantification of ER-mitochondria signaling proteins by immunoblotting revealed loss of VAPB and PTPIP51 in cortex but not cerebellum at end-stage Alzheimer's disease. Proximity ligation assays were used to quantify the VAPB-PTPIP51 interaction in temporal cortex pyramidal neurons and cerebellar Purkinje cell neurons in control, Braak stage III-IV (early/mid-dementia) and Braak stage VI (severe dementia) cases. Pyramidal neurons degenerate in Alzheimer's disease whereas Purkinje cells are less affected. These studies revealed that the VAPB-PTPIP51 tethers are disrupted in Braak stage III-IV pyramidal but not Purkinje cell neurons. Thus, we identify a new pathogenic event in post-mortem Alzheimer's disease brains. The implications of our findings for Alzheimer's disease mechanisms are discussed.


Subject(s)
Alzheimer Disease/metabolism , Endoplasmic Reticulum/metabolism , Mitochondria/metabolism , Mitochondrial Proteins/metabolism , Protein Tyrosine Phosphatases/metabolism , Temporal Lobe/metabolism , Vesicular Transport Proteins/metabolism , Aged , Aged, 80 and over , Alzheimer Disease/pathology , Autopsy , Endoplasmic Reticulum/pathology , Female , Humans , Male , Mitochondria/pathology , Pyramidal Cells/metabolism , Pyramidal Cells/pathology , Temporal Lobe/pathology
9.
Acta Neuropathol Commun ; 7(1): 35, 2019 03 06.
Article in English | MEDLINE | ID: mdl-30841933

ABSTRACT

Signaling between the endoplasmic reticulum (ER) and mitochondria regulates a number of key neuronal functions. This signaling involves close physical contacts between the two organelles that are mediated by "tethering proteins" that function to recruit regions of ER to the mitochondrial surface. The ER protein, vesicle-associated membrane protein-associated protein B (VAPB) and the mitochondrial membrane protein, protein tyrosine phosphatase interacting protein-51 (PTPIP51), interact to form one such tether. Recently, damage to ER-mitochondria signaling involving disruption of the VAPB-PTPIP51 tethers has been linked to the pathogenic process in Parkinson's disease, fronto-temporal dementia (FTD) and related amyotrophic lateral sclerosis (ALS). Loss of neuronal synaptic function is a key feature of Parkinson's disease and FTD/ALS but the roles that ER-mitochondria signaling and the VAPB-PTPIP51 tethers play in synaptic function are not known. Here, we demonstrate that the VAPB-PTPIP51 tethers regulate synaptic activity. VAPB and PTPIP51 localise and form contacts at synapses, and stimulating neuronal activity increases ER-mitochondria contacts and the VAPB-PTPIP51 interaction. Moreover, siRNA loss of VAPB or PTPIP51 perturbs synaptic function and dendritic spine morphology. Our results reveal a new role for the VAPB-PTPIP51 tethers in neurons and suggest that damage to ER-mitochondria signaling contributes to synaptic dysfunction in Parkinson's disease and FTD/ALS.


Subject(s)
Endoplasmic Reticulum/metabolism , Kv Channel-Interacting Proteins/metabolism , Mitochondrial Proteins/metabolism , Neurons/metabolism , Protein Tyrosine Phosphatases/metabolism , Synapses/metabolism , Animals , Cells, Cultured , Endoplasmic Reticulum/chemistry , Hippocampus/chemistry , Hippocampus/metabolism , Kv Channel-Interacting Proteins/analysis , Mitochondrial Proteins/analysis , Neurons/chemistry , Protein Tyrosine Phosphatases/analysis , Rats , Synapses/chemistry
10.
Acta Neuropathol Commun ; 6(1): 65, 2018 07 23.
Article in English | MEDLINE | ID: mdl-30037345

ABSTRACT

Tau is normally a highly soluble phosphoprotein found predominantly in neurons. Six different isoforms of tau are expressed in the adult human CNS. Under pathological conditions, phosphorylated tau aggregates are a defining feature of neurodegenerative disorders called tauopathies. Recent findings have suggested a potential role of the gut-brain axis in CNS homeostasis, and therefore we set out to examine the isoform profile and phosphorylation state of tau in the enteric nervous system (ENS) under physiological conditions and in tauopathies. Surgical specimens of human colon from controls, Parkinson's disease (PD) and progressive supranuclear palsy (PSP) patients were analyzed by Western Blot and immunohistochemistry using a panel of anti-tau antibodies. We found that adult human ENS primarily expresses two tau isoforms, localized in the cell bodies and neuronal processes. We did not observe any difference in the enteric tau isoform profile and phosphorylation state between PSP, PD and control subjects. The htau mouse model of tauopathy also expressed two main isoforms of human tau in the ENS, and there were no apparent differences in ENS tau localization or phosphorylation between wild-type and htau mice. Tau in both human and mouse ENS was found to be phosphorylated but poorly susceptible to dephosphorylation with lambda phosphatase. To investigate ENS tau phosphorylation further, primary cultures from rat enteric neurons, which express four isoforms of tau, were pharmacologically manipulated to show that ENS tau phosphorylation state can be regulated, at least in vitro. Our study is the first to characterize tau in the rodent and human ENS. As a whole, our findings provide a basis to unravel the functions of tau in the ENS and to further investigate the possibility of pathological changes in enteric neuropathies and tauopathies.


Subject(s)
Enteric Nervous System/metabolism , Parkinson Disease/pathology , Supranuclear Palsy, Progressive/pathology , tau Proteins/metabolism , Aged , Animals , Anti-Infective Agents/pharmacology , Benzophenanthridines/pharmacology , Brain/metabolism , Brain/pathology , Cells, Cultured , Colon/metabolism , Colon/pathology , Embryo, Mammalian , Enteric Nervous System/drug effects , Female , Humans , Isoquinolines/pharmacology , Male , Mice , Mice, Transgenic , Middle Aged , Myenteric Plexus/metabolism , Pregnancy , Protein Isoforms/genetics , Protein Isoforms/metabolism , Rats , Rats, Sprague-Dawley , Submucous Plexus/metabolism , Tubulin/metabolism , Ubiquitin Thiolesterase/metabolism , Young Adult , tau Proteins/genetics
11.
Sci Rep ; 8(1): 9033, 2018 06 13.
Article in English | MEDLINE | ID: mdl-29899557

ABSTRACT

Mitochondrial dysfunction is implicated in many neurodegenerative diseases including Parkinson's disease (PD). Induced pluripotent stem cells (iPSCs) provide a unique cell model for studying neurological diseases. We have established a high-content assay that can simultaneously measure mitochondrial function, morphology and cell viability in iPSC-derived dopaminergic neurons. iPSCs from PD patients with mutations in SNCA and unaffected controls were differentiated into dopaminergic neurons, seeded in 384-well plates and stained with the mitochondrial membrane potential dependent dye TMRM, alongside Hoechst-33342 and Calcein-AM. Images were acquired using an automated confocal screening microscope and single cells were analysed using automated image analysis software. PD neurons displayed reduced mitochondrial membrane potential and altered mitochondrial morphology compared to control neurons. This assay demonstrates that high content screening techniques can be applied to the analysis of mitochondria in iPSC-derived neurons. This technique could form part of a drug discovery platform to test potential new therapeutics for PD and other neurodegenerative diseases.


Subject(s)
Dopaminergic Neurons/metabolism , Induced Pluripotent Stem Cells/metabolism , Mitochondria/metabolism , Mutation , Single-Cell Analysis/methods , alpha-Synuclein/metabolism , Benzimidazoles , Cell Differentiation/genetics , Cell Survival/genetics , Cells, Cultured , Humans , Membrane Potential, Mitochondrial/genetics , Microscopy, Confocal , Mitochondria/genetics , Parkinson Disease/genetics , Parkinson Disease/metabolism , Parkinson Disease/pathology , Rhodamines , alpha-Synuclein/genetics
12.
Cell Death Dis ; 9(3): 327, 2018 02 28.
Article in English | MEDLINE | ID: mdl-29491392

ABSTRACT

Fronto-temporal dementia (FTD) and amyotrophic lateral sclerosis (ALS) are two related and incurable neurodegenerative diseases. Features of these diseases include pathological protein inclusions in affected neurons with TAR DNA-binding protein 43 (TDP-43), dipeptide repeat proteins derived from the C9ORF72 gene, and fused in sarcoma (FUS) representing major constituent proteins in these inclusions. Mutations in C9ORF72 and the genes encoding TDP-43 and FUS cause familial forms of FTD/ALS which provides evidence to link the pathology and genetics of these diseases. A large number of seemingly disparate physiological functions are damaged in FTD/ALS. However, many of these damaged functions are regulated by signalling between the endoplasmic reticulum and mitochondria, and this has stimulated investigations into the role of endoplasmic reticulum-mitochondria signalling in FTD/ALS disease processes. Here, we review progress on this topic.


Subject(s)
Amyotrophic Lateral Sclerosis/metabolism , Dementia/metabolism , Endoplasmic Reticulum/metabolism , Mitochondria/metabolism , Amyotrophic Lateral Sclerosis/genetics , Animals , C9orf72 Protein/genetics , C9orf72 Protein/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Dementia/genetics , Endoplasmic Reticulum/genetics , Humans , Mitochondria/genetics , Signal Transduction
13.
Autophagy ; 13(7): 1250-1251, 2017 Jul 03.
Article in English | MEDLINE | ID: mdl-28548902

ABSTRACT

The endoplasmic reticulum (ER) and mitochondria form tight functional contacts that regulate several key cellular processes. The formation of these contacts involves "tethering proteins" that function to recruit regions of ER to mitochondria. The integral ER protein VAPB (VAMP associated protein B and C) binds to the outer mitochondrial membrane protein, RMDN3/PTPIP51 (regulator of microtubule dynamics 3) to form one such set of tethers. Recently, we showed that the VAPB-RMDN3 tethers regulate macroautophagy/autophagy. Small interfering RNA (siRNA) knockdown of VAPB or RMDN3 to loosen ER-mitochondria contacts stimulates autophagosome formation, whereas overexpression of VAPB or RMDN3 to tighten contacts inhibit their formation. Artificial tethering of ER and mitochondria via expression of a synthetic linker protein also reduces autophagy and this artificial tether rescues the effects of VAPB- or RMDN3-targeted siRNA loss on autophagosome formation. Finally, our studies revealed that the modulatory effects of ER-mitochondria contacts on autophagy involve their role in mediating ITPR (inositol 1,4,5-trisphosphate receptor) delivery of Ca2+ from ER stores to mitochondria.


Subject(s)
Autophagy , Endoplasmic Reticulum/metabolism , Mitochondria/metabolism , Calcium/metabolism , Membrane Proteins/metabolism , Mitochondrial Proteins/metabolism , Signal Transduction
14.
Acta Neuropathol ; 134(1): 129-149, 2017 07.
Article in English | MEDLINE | ID: mdl-28337542

ABSTRACT

α-Synuclein is strongly linked to Parkinson's disease but the molecular targets for its toxicity are not fully clear. However, many neuronal functions damaged in Parkinson's disease are regulated by signalling between the endoplasmic reticulum (ER) and mitochondria. This signalling involves close physical associations between the two organelles that are mediated by binding of the integral ER protein vesicle-associated membrane protein-associated protein B (VAPB) to the outer mitochondrial membrane protein, protein tyrosine phosphatase-interacting protein 51 (PTPIP51). VAPB and PTPIP51 thus act as a scaffold to tether the two organelles. Here we show that α-synuclein binds to VAPB and that overexpression of wild-type and familial Parkinson's disease mutant α-synuclein disrupt the VAPB-PTPIP51 tethers to loosen ER-mitochondria associations. This disruption to the VAPB-PTPIP51 tethers is also seen in neurons derived from induced pluripotent stem cells from familial Parkinson's disease patients harbouring pathogenic triplication of the α-synuclein gene. We also show that the α-synuclein induced loosening of ER-mitochondria contacts is accompanied by disruption to Ca2+ exchange between the two organelles and mitochondrial ATP production. Such disruptions are likely to be particularly damaging to neurons that are heavily dependent on correct Ca2+ signaling and ATP.


Subject(s)
Adenosine Triphosphate/metabolism , Calcium/metabolism , Homeostasis/physiology , Mitochondria/metabolism , Vesicular Transport Proteins/metabolism , alpha-Synuclein/metabolism , Animals , Cations, Divalent/metabolism , Cell Line, Tumor , Dopaminergic Neurons/metabolism , Dopaminergic Neurons/pathology , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum/pathology , Glycogen Synthase Kinase 3 beta/metabolism , HEK293 Cells , Humans , Induced Pluripotent Stem Cells/metabolism , Induced Pluripotent Stem Cells/pathology , Mitochondria/pathology , Mitochondrial Proteins/metabolism , Mutation , Parkinson Disease/genetics , Parkinson Disease/metabolism , Parkinson Disease/pathology , Protein Tyrosine Phosphatases/metabolism , Rats, Sprague-Dawley , alpha-Synuclein/genetics
15.
Curr Biol ; 27(3): 371-385, 2017 Feb 06.
Article in English | MEDLINE | ID: mdl-28132811

ABSTRACT

Mitochondria form close physical associations with the endoplasmic reticulum (ER) that regulate a number of physiological functions. One mechanism by which regions of ER are recruited to mitochondria involves binding of the ER protein VAPB to the mitochondrial protein PTPIP51, which act as scaffolds to tether the two organelles. Here, we show that the VAPB-PTPIP51 tethers regulate autophagy. We demonstrate that overexpression of VAPB or PTPIP51 to tighten ER-mitochondria contacts impairs, whereas small interfering RNA (siRNA)-mediated loss of VAPB or PTPIP51 to loosen contacts stimulates, autophagosome formation. Moreover, we show that expression of a synthetic linker protein that artificially tethers ER and mitochondria also reduces autophagosome formation, and that this artificial tether rescues the effects of siRNA loss of VAPB or PTPIP51 on autophagy. Thus, these effects of VAPB and PTPIP51 manipulation on autophagy are a consequence of their ER-mitochondria tethering function. Interestingly, we discovered that tightening of ER-mitochondria contacts by overexpression of VAPB or PTPIP51 impairs rapamycin- and torin 1-induced, but not starvation-induced, autophagy. This suggests that the regulation of autophagy by ER-mitochondria signaling is at least partly dependent upon the nature of the autophagic stimulus. Finally, we demonstrate that the mechanism by which the VAPB-PTPIP51 tethers regulate autophagy involves their role in mediating delivery of Ca2+ to mitochondria from ER stores. Thus, our findings reveal a new molecular mechanism for regulating autophagy.


Subject(s)
Autophagy , Endoplasmic Reticulum/metabolism , Mitochondria/metabolism , Mitochondrial Proteins/metabolism , Protein Tyrosine Phosphatases/metabolism , Vesicular Transport Proteins/metabolism , Blood Proteins/pharmacology , Calcium/metabolism , Endoplasmic Reticulum/drug effects , Gene Expression Regulation/drug effects , HEK293 Cells , Humans , Immunosuppressive Agents/pharmacology , Mitochondria/drug effects , Mitochondrial Proteins/antagonists & inhibitors , Mitochondrial Proteins/genetics , Protein Tyrosine Phosphatases/antagonists & inhibitors , Protein Tyrosine Phosphatases/genetics , RNA, Small Interfering/genetics , Sirolimus/pharmacology , Starvation , Vesicular Transport Proteins/antagonists & inhibitors , Vesicular Transport Proteins/genetics
16.
EMBO Rep ; 17(9): 1326-42, 2016 09.
Article in English | MEDLINE | ID: mdl-27418313

ABSTRACT

Defective FUS metabolism is strongly associated with amyotrophic lateral sclerosis and frontotemporal dementia (ALS/FTD), but the mechanisms linking FUS to disease are not properly understood. However, many of the functions disrupted in ALS/FTD are regulated by signalling between the endoplasmic reticulum (ER) and mitochondria. This signalling is facilitated by close physical associations between the two organelles that are mediated by binding of the integral ER protein VAPB to the outer mitochondrial membrane protein PTPIP51, which act as molecular scaffolds to tether the two organelles. Here, we show that FUS disrupts the VAPB-PTPIP51 interaction and ER-mitochondria associations. These disruptions are accompanied by perturbation of Ca(2+) uptake by mitochondria following its release from ER stores, which is a physiological read-out of ER-mitochondria contacts. We also demonstrate that mitochondrial ATP production is impaired in FUS-expressing cells; mitochondrial ATP production is linked to Ca(2+) levels. Finally, we demonstrate that the FUS-induced reductions to ER-mitochondria associations and are linked to activation of glycogen synthase kinase-3ß (GSK-3ß), a kinase already strongly associated with ALS/FTD.


Subject(s)
Amyotrophic Lateral Sclerosis/metabolism , Endoplasmic Reticulum/metabolism , Frontotemporal Dementia/metabolism , Glycogen Synthase Kinase 3 beta/metabolism , Mitochondria/metabolism , Mitochondrial Proteins/metabolism , Protein Tyrosine Phosphatases/metabolism , RNA-Binding Protein FUS/metabolism , Vesicular Transport Proteins/metabolism , Adenosine Triphosphate/metabolism , Animals , Calcium/metabolism , Disease Models, Animal , Endoplasmic Reticulum/ultrastructure , Enzyme Activation , Gene Expression , Humans , Mice , Mice, Transgenic , Mitochondria/ultrastructure , Mutation , Protein Binding , RNA-Binding Protein FUS/genetics
17.
Trends Neurosci ; 39(3): 146-157, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26899735

ABSTRACT

Alzheimer's disease (AD), Parkinson's disease (PD), and amyotrophic lateral sclerosis with associated frontotemporal dementia (ALS/FTD) are major neurodegenerative diseases for which there are no cures. All are characterised by damage to several seemingly disparate cellular processes. The broad nature of this damage makes understanding pathogenic mechanisms and devising new treatments difficult. Can the different damaged functions be linked together in a common disease pathway and which damaged function should be targeted for therapy? Many functions damaged in neurodegenerative diseases are regulated by communications that mitochondria make with a specialised region of the endoplasmic reticulum (ER; mitochondria-associated ER membranes or 'MAM'). Moreover, several recent studies have shown that disturbances to ER-mitochondria contacts occur in neurodegenerative diseases. Here, we review these findings.


Subject(s)
Endoplasmic Reticulum/metabolism , Mitochondria/metabolism , Neurodegenerative Diseases/metabolism , Animals , Endoplasmic Reticulum/ultrastructure , Humans , Mitochondria/ultrastructure , Neurodegenerative Diseases/genetics , Neurodegenerative Diseases/pathology , Neurodegenerative Diseases/therapy
18.
Nat Commun ; 5: 3996, 2014 Jun 03.
Article in English | MEDLINE | ID: mdl-24893131

ABSTRACT

Mitochondria and the endoplasmic reticulum (ER) form tight structural associations and these facilitate a number of cellular functions. However, the mechanisms by which regions of the ER become tethered to mitochondria are not properly known. Understanding these mechanisms is not just important for comprehending fundamental physiological processes but also for understanding pathogenic processes in some disease states. In particular, disruption to ER-mitochondria associations is linked to some neurodegenerative diseases. Here we show that the ER-resident protein VAPB interacts with the mitochondrial protein tyrosine phosphatase-interacting protein-51 (PTPIP51) to regulate ER-mitochondria associations. Moreover, we demonstrate that TDP-43, a protein pathologically linked to amyotrophic lateral sclerosis and fronto-temporal dementia perturbs ER-mitochondria interactions and that this is associated with disruption to the VAPB-PTPIP51 interaction and cellular Ca(2+) homeostasis. Finally, we show that overexpression of TDP-43 leads to activation of glycogen synthase kinase-3ß (GSK-3ß) and that GSK-3ß regulates the VAPB-PTPIP51 interaction. Our results describe a new pathogenic mechanism for TDP-43.


Subject(s)
DNA-Binding Proteins/metabolism , Endoplasmic Reticulum/metabolism , Glycogen Synthase Kinase 3/metabolism , Mitochondria/metabolism , Mitochondrial Proteins/metabolism , Protein Tyrosine Phosphatases/metabolism , Vesicular Transport Proteins/metabolism , Amyotrophic Lateral Sclerosis/metabolism , Animals , Calcium/metabolism , Cell Line , Frontotemporal Dementia/metabolism , Glycogen Synthase Kinase 3 beta , HEK293 Cells , Humans , Membrane Proteins/metabolism , Mice
19.
J Neurochem ; 125(4): 512-7, 2013 May.
Article in English | MEDLINE | ID: mdl-23278133

ABSTRACT

There is growing evidence supporting a role of extracellular alpha-synuclein in the spreading of Parkinson's disease (PD) pathology. Recent pathological studies have raised the possibility that the enteric nervous system (ENS) is one of the initial sites of alpha-synuclein pathology in PD. We therefore undertook this survey to determine whether alpha-synuclein can be secreted by enteric neurons. Alpha-synuclein secretion was assessed by immunoblot analysis of the culture medium from primary culture of ENS. We show that alpha-synuclein is physiologically secreted by enteric neurons via a conventional, endoplasmic reticulum/Golgi-dependent exocytosis, in a neuronal activity-regulated manner. Our study is the first to evidence that enteric neurons are capable of secreting alpha-synuclein, thereby providing new insights into the role of the ENS in the pathophysiology of PD.


Subject(s)
Enteric Nervous System/metabolism , Intestine, Small/innervation , Neurons/metabolism , Parkinson Disease/metabolism , alpha-Synuclein/metabolism , Animals , Blotting, Western , Brefeldin A/pharmacology , Colforsin/pharmacology , Enteric Nervous System/cytology , Exocytosis/physiology , Neurons/cytology , Neurons/drug effects , Parkinson Disease/pathology , Parkinson Disease/physiopathology , Primary Cell Culture , Protein Synthesis Inhibitors/pharmacology , Rats
20.
PLoS One ; 5(9): e12728, 2010 Sep 14.
Article in English | MEDLINE | ID: mdl-20856865

ABSTRACT

BACKGROUND: The presence of Lewy bodies and Lewy neurites (LN) has been demonstrated in the enteric nervous system (ENS) of Parkinson's disease (PD) patients. The aims of the present research were to use routine colonoscopy biopsies (1) to analyze, in depth, enteric pathology throughout the colonic submucosal plexus (SMP), and (2) to correlate the pathological burden with neurological and gastrointestinal (GI) symptoms. METHODOLOGY/PRINCIPAL FINDINGS: A total of 10 control and 29 PD patients divided into 3 groups according to disease duration were included. PD and GI symptoms were assessed using the Unified Parkinson's Disease Rating Scale part III and the Rome III questionnaire, respectively. Four biopsies were taken from the ascending and descending colon during the course of a total colonoscopy. Immunohistochemical analysis was performed using antibodies against phosphorylated alpha-synuclein, neurofilaments NF 220 kDa (NF) and tyrosine hydroxylase (TH). The density of LN, labeled by anti-phosphorylated alpha-synuclein antibodies, was evaluated using a quantitative rating score. Lewy pathology was apparent in the colonic biopsies from 21 patients and in none of the controls. A decreased number of NF-immunoreactive neurons per ganglion was observed in the SMP of PD patients compared to controls. The amount of LN in the ENS was inversely correlated with neuronal count and positively correlated with levodopa-unresponsive features and constipation. CONCLUSION/SIGNIFICANCE: Analysis of the ENS by routine colonoscopy biopsies is a useful tool for pre-mortem neuropathological diagnosis of PD, and also provides insight into the progression of motor and non-motor symptoms.


Subject(s)
Colon/innervation , Colon/pathology , Parkinson Disease/diagnosis , Parkinson Disease/pathology , Adult , Aged , Biopsy , Colon/metabolism , Colonoscopy , Enteric Nervous System/metabolism , Enteric Nervous System/pathology , Female , Humans , Lewy Bodies/metabolism , Male , Middle Aged , Parkinson Disease/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL