51
|
The role of mitochondria-associated membranes in cellular homeostasis and diseases. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2019; 350:119-196. [PMID: 32138899 DOI: 10.1016/bs.ircmb.2019.11.002] [Citation(s) in RCA: 83] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Mitochondria and endoplasmic reticulum (ER) are fundamental in the control of cell physiology regulating several signal transduction pathways. They continuously communicate exchanging messages in their contact sites called MAMs (mitochondria-associated membranes). MAMs are specific microdomains acting as a platform for the sorting of vital and dangerous signals. In recent years increasing evidence reported that multiple scaffold proteins and regulatory factors localize to this subcellular fraction suggesting MAMs as hotspot signaling domains. In this review we describe the current knowledge about MAMs' dynamics and processes, which provided new correlations between MAMs' dysfunctions and human diseases. In fact, MAMs machinery is strictly connected with several pathologies, like neurodegeneration, diabetes and mainly cancer. These pathological events are characterized by alterations in the normal communication between ER and mitochondria, leading to deep metabolic defects that contribute to the progression of the diseases.
Collapse
|
52
|
Abraham MJ, Fleming KL, Raymond S, Wong AYC, Bergeron R. The sigma-1 receptor behaves as an atypical auxiliary subunit to modulate the functional characteristics of Kv1.2 channels expressed in HEK293 cells. Physiol Rep 2019; 7:e14147. [PMID: 31222975 PMCID: PMC6586770 DOI: 10.14814/phy2.14147] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Revised: 05/17/2019] [Accepted: 05/25/2019] [Indexed: 12/11/2022] Open
Abstract
Expression of Kv1.2 within Kv1.x potassium channel complexes is critical in maintaining appropriate neuronal excitability and determining the threshold for action potential firing. This is attributed to the interaction of Kv1.2 with a hitherto unidentified protein that confers bimodal channel activation gating, allowing neurons to adapt to repetitive trains of stimulation and protecting against hyperexcitability. One potential protein candidate is the sigma-1 receptor (Sig-1R), which regulates other members of the Kv1.x channel family; however, the biophysical nature of the interaction between Sig-1R and Kv1.2 has not been elucidated. We hypothesized that Sig-1R may regulate Kv1.2 and may further act as the unidentified modulator of Kv1.2 activation. In transiently transfected HEK293 cells, we found that ligand activation of the Sig-1R modulates Kv1.2 current amplitude. More importantly, Sig-1R interacts with Kv1.2 in baseline conditions to influence bimodal activation gating. These effects are abolished in the presence of the auxiliary subunit Kvβ2 and when the Sig-1R mutation underlying ALS16 (Sig-1R-E102Q), is expressed. These data suggest that Kvβ2 occludes the interaction of Sig-1R with Kv1.2, and that E102 may be a residue critical for Sig-1R modulation of Kv1.2. The results of this investigation describe an important new role for Sig-1R in the regulation of neuronal excitability and introduce a novel mechanism of pathophysiology in Sig-1R dysfunction.
Collapse
Affiliation(s)
- Madelyn J. Abraham
- Department of Cellular and Molecular MedicineUniversity of OttawaOttawaOntarioCanada
| | - Kayla L. Fleming
- Department of Cellular and Molecular MedicineUniversity of OttawaOttawaOntarioCanada
| | - Sophie Raymond
- NeuroscienceOttawa Hospital Research InstituteOttawaOntarioCanada
| | | | - Richard Bergeron
- Department of Cellular and Molecular MedicineUniversity of OttawaOttawaOntarioCanada
- NeuroscienceOttawa Hospital Research InstituteOttawaOntarioCanada
| |
Collapse
|
53
|
Francardo V, Geva M, Bez F, Denis Q, Steiner L, Hayden MR, Cenci MA. Pridopidine Induces Functional Neurorestoration Via the Sigma-1 Receptor in a Mouse Model of Parkinson's Disease. Neurotherapeutics 2019; 16:465-479. [PMID: 30756361 PMCID: PMC6554374 DOI: 10.1007/s13311-018-00699-9] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Pridopidine is a small molecule in clinical development for the treatment of Huntington's disease. It was recently found to have high binding affinity to the sigma-1 receptor, a chaperone protein involved in cellular defense mechanisms and neuroplasticity. Here, we have evaluated the neuroprotective and neurorestorative effects of pridopidine in a unilateral 6-hydroxydopamine (6-OHDA) lesion model of parkinsonism in mice. By 5 weeks of daily administration, a low dose of pridopidine (0.3 mg/kg) had significantly improved deficits in forelimb use (cylinder test, stepping test) and abolished the ipsilateral rotational bias typical of hemiparkinsonian animals. A higher dose of pridopidine (1 mg/kg) significantly improved only the rotational bias, with a trend towards improvement in forelimb use. The behavioral recovery induced by pridopidine 0.3 mg/kg was accompanied by a significant protection of nigral dopamine cell bodies, an increased dopaminergic fiber density in the striatum, and striatal upregulation of GDNF, BDNF, and phosphorylated ERK1/2. The beneficial effects of pridopidine 0.3 mg/kg were absent in 6-OHDA-lesioned mice lacking the sigma-1 receptor. Pharmacokinetic data confirmed that the effective dose of pridopidine reached brain concentrations sufficient to bind S1R. Our results are the first to show that pridopidine promotes functional neurorestoration in the damaged nigrostriatal system acting via the sigma-1 receptor.
Collapse
Affiliation(s)
- Veronica Francardo
- Basal Ganglia Pathophysiology Unit, Department of Experimental Medical Science, Lund University, BMC F11, Lund, Sweden
| | | | - Francesco Bez
- Basal Ganglia Pathophysiology Unit, Department of Experimental Medical Science, Lund University, BMC F11, Lund, Sweden
| | - Quentin Denis
- Basal Ganglia Pathophysiology Unit, Department of Experimental Medical Science, Lund University, BMC F11, Lund, Sweden
| | - Lilach Steiner
- Teva Pharmaceutical Industries Global Research and Development, Netanya, Israel
| | | | - M Angela Cenci
- Basal Ganglia Pathophysiology Unit, Department of Experimental Medical Science, Lund University, BMC F11, Lund, Sweden.
| |
Collapse
|
54
|
Chaplot K, Pimpale L, Ramalingam B, Deivasigamani S, Kamat SS, Ratnaparkhi GS. SOD1 activity threshold and TOR signalling modulate VAP(P58S) aggregation via reactive oxygen species-induced proteasomal degradation in a Drosophila model of amyotrophic lateral sclerosis. Dis Model Mech 2019; 12:dmm.033803. [PMID: 30635270 PMCID: PMC6398501 DOI: 10.1242/dmm.033803] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2018] [Accepted: 01/07/2019] [Indexed: 12/11/2022] Open
Abstract
Familial amyotrophic lateral sclerosis (ALS) is an incurable, late-onset motor neuron disease, linked strongly to various causative genetic loci. ALS8 codes for a missense mutation, P56S, in VAMP-associated protein B (VAPB) that causes the protein to misfold and form cellular aggregates. Uncovering genes and mechanisms that affect aggregation dynamics would greatly help increase our understanding of the disease and lead to potential therapeutics. We developed a quantitative high-throughput Drosophila S2R+ cell-based kinetic assay coupled with fluorescent microscopy to score for genes involved in the modulation of aggregates of the fly orthologue, VAP(P58S), fused with GFP. A targeted RNA interference screen against 900 genes identified 150 hits that modify aggregation, including the ALS loci Sod1 and TDP43 (also known as TBPH), as well as genes belonging to the mTOR pathway. Further, a system to measure the extent of VAP(P58S) aggregation in the Drosophila larval brain was developed in order to validate the hits from the cell-based screen. In the larval brain, we find that reduction of SOD1 levels or decreased mTOR signalling reduces aggregation, presumably by increasing the levels of cellular reactive oxygen species (ROS). The mechanism of aggregate clearance is, primarily, proteasomal degradation, which appears to be triggered by an increase in ROS. We have thus uncovered an interesting interplay between SOD1, ROS and mTOR signalling that regulates the dynamics of VAP aggregation. Mechanistic processes underlying such cellular regulatory networks will lead to better understanding of the initiation and progression of ALS.This article has an associated First Person interview with the first author of the paper.
Collapse
Affiliation(s)
- Kriti Chaplot
- Department of Biology, Indian Institute of Science Education and Research, Pune 411008, India
| | - Lokesh Pimpale
- Department of Biology, Indian Institute of Science Education and Research, Pune 411008, India
| | | | | | - Siddhesh S Kamat
- Department of Biology, Indian Institute of Science Education and Research, Pune 411008, India
| | - Girish S Ratnaparkhi
- Department of Biology, Indian Institute of Science Education and Research, Pune 411008, India
| |
Collapse
|
55
|
Saito A, Imaizumi K. Unfolded Protein Response-Dependent Communication and Contact among Endoplasmic Reticulum, Mitochondria, and Plasma Membrane. Int J Mol Sci 2018; 19:ijms19103215. [PMID: 30340324 PMCID: PMC6213962 DOI: 10.3390/ijms19103215] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Revised: 10/10/2018] [Accepted: 10/13/2018] [Indexed: 12/20/2022] Open
Abstract
The function of the endoplasmic reticulum (ER) can be impaired by changes to the extra- and intracellular environment, such as disruption of calcium homeostasis, expression of mutated proteins, and oxidative stress. In response to disruptions to ER homeostasis, eukaryotic cells activate canonical branches of signal transduction cascades, collectively termed the unfolded protein response (UPR). The UPR functions to remove or recover the activity of misfolded proteins that accumulated in the ER and to avoid irreversible cellular damage. Additionally, the UPR plays unique physiological roles in the regulation of diverse cellular events, including cell differentiation and development and lipid biosynthesis. Recent studies have shown that these important cellular events are also regulated by contact and communication among organelles. These reports suggest strong involvement among the UPR, organelle communication, and regulation of cellular homeostasis. However, the precise mechanisms for the formation of contact sites and the regulation of ER dynamics by the UPR remain unresolved. In this review, we summarize the current understanding of how the UPR regulates morphological changes to the ER and the formation of contact sites between the ER and other organelles. We also review how UPR-dependent connections between the ER and other organelles affect cellular and physiological functions.
Collapse
Affiliation(s)
- Atsushi Saito
- Department of Stress Protein Processing, Institute of Biomedical & Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima 734-8553, Japan.
| | - Kazunori Imaizumi
- Department of Biochemistry, Institute of Biomedical & Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima 734-8553, Japan.
| |
Collapse
|
56
|
|
57
|
Abdullah CS, Alam S, Aishwarya R, Miriyala S, Panchatcharam M, Bhuiyan MAN, Peretik JM, Orr AW, James J, Osinska H, Robbins J, Lorenz JN, Bhuiyan MS. Cardiac Dysfunction in the Sigma 1 Receptor Knockout Mouse Associated With Impaired Mitochondrial Dynamics and Bioenergetics. J Am Heart Assoc 2018; 7:e009775. [PMID: 30371279 PMCID: PMC6474981 DOI: 10.1161/jaha.118.009775] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Accepted: 09/10/2018] [Indexed: 12/21/2022]
Abstract
Background The Sigma 1 receptor (Sigmar1) functions as an interorganelle signaling molecule and elicits cytoprotective functions. The presence of Sigmar1 in the heart was first reported on the basis of a ligand-binding assay, and all studies to date have been limited to pharmacological approaches using less-selective ligands for Sigmar1. However, the physiological function of cardiac Sigmar1 remains unknown. We investigated the physiological function of Sigmar1 in regulating cardiac hemodynamics using the Sigmar1 knockout mouse (Sigmar1-/-). Methods and Results Sigmar1-/- hearts at 3 to 4 months of age showed significantly increased contractility as assessed by left ventricular catheterization with stimulation by increasing doses of a β1-adrenoceptor agonist. Noninvasive echocardiographic measurements were also used to measure cardiac function over time, and the data showed the development of cardiac contractile dysfunction in Sigmar1 -/- hearts as the animals aged. Histochemistry demonstrated significant cardiac fibrosis, collagen deposition, and increased periostin in the Sigmar1 -/- hearts compared with wild-type hearts. Ultrastructural analysis of Sigmar1-/- cardiomyocytes revealed an irregularly shaped, highly fused mitochondrial network with abnormal cristae. Mitochondrial size was larger in Sigmar1-/- hearts, resulting in decreased numbers of mitochondria per microscopic field. In addition, Sigmar1-/- hearts showed altered expression of mitochondrial dynamics regulatory proteins. Real-time oxygen consumption rates in isolated mitochondria showed reduced respiratory function in Sigmar1-/- hearts compared with wild-type hearts. Conclusions We demonstrate a potential function of Sigmar1 in regulating normal mitochondrial organization and size in the heart. Sigmar1 loss of function led to mitochondrial dysfunction, abnormal mitochondrial architecture, and adverse cardiac remodeling, culminating in cardiac contractile dysfunction.
Collapse
Affiliation(s)
- Chowdhury S. Abdullah
- Department of Pathology and Translational PathobiologyLouisiana State University Health Sciences CenterShreveportLA
| | - Shafiul Alam
- Department of Pathology and Translational PathobiologyLouisiana State University Health Sciences CenterShreveportLA
| | - Richa Aishwarya
- Department of Molecular and Cellular PhysiologyLouisiana State University Health Sciences CenterShreveportLA
| | - Sumitra Miriyala
- Department of Cellular Biology and AnatomyLouisiana State University Health Sciences CenterShreveportLA
| | - Manikandan Panchatcharam
- Department of Cellular Biology and AnatomyLouisiana State University Health Sciences CenterShreveportLA
| | | | - Jonette M. Peretik
- Department of Pathology and Translational PathobiologyLouisiana State University Health Sciences CenterShreveportLA
| | - A. Wayne Orr
- Department of Pathology and Translational PathobiologyLouisiana State University Health Sciences CenterShreveportLA
- Department of Molecular and Cellular PhysiologyLouisiana State University Health Sciences CenterShreveportLA
- Department of Cellular Biology and AnatomyLouisiana State University Health Sciences CenterShreveportLA
| | - Jeanne James
- Division of Pediatric CardiologyMedical College of WisconsinMilwaukeeWI
| | - Hanna Osinska
- Division of Molecular Cardiovascular BiologyCincinnati Children's HospitalCincinnatiOH
| | - Jeffrey Robbins
- Division of Molecular Cardiovascular BiologyCincinnati Children's HospitalCincinnatiOH
| | - John N. Lorenz
- Department of Molecular and Cellular PhysiologyUniversity of Cincinnati College of MedicineCincinnatiOH
| | - Md. Shenuarin Bhuiyan
- Department of Pathology and Translational PathobiologyLouisiana State University Health Sciences CenterShreveportLA
- Department of Molecular and Cellular PhysiologyLouisiana State University Health Sciences CenterShreveportLA
| |
Collapse
|
58
|
Jia J, Cheng J, Wang C, Zhen X. Sigma-1 Receptor-Modulated Neuroinflammation in Neurological Diseases. Front Cell Neurosci 2018; 12:314. [PMID: 30294261 PMCID: PMC6158303 DOI: 10.3389/fncel.2018.00314] [Citation(s) in RCA: 70] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Accepted: 08/29/2018] [Indexed: 02/02/2023] Open
Abstract
A large body of evidence indicates that sigma-1 receptors (Sig-1R) are important drug targets for a number of neuropsychiatric disorders. Sig-1Rs are enriched in central nervous system (CNS). In addition to neurons, both cerebral microglia and astrocytes express Sig-1Rs. Activation of Sig-1Rs is known to elicit potent neuroprotective effects and promote neuronal survival via multiple mechanisms, including promoting mitochondrial functions, decreasing oxidative stress and regulating neuroimmnological functions. In this review article, we focus on the emerging role of Sig-1Rs in regulating neuroinflammation and discuss the recent advances on the Sig-1R-modulating neuroinflammation in the pathophysiology and therapy of neurodegenerative disorders.
Collapse
Affiliation(s)
- Jia Jia
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| | - Jian Cheng
- Institute of Neuroscience, Soochow University, Suzhou, China
| | - Cheng Wang
- Department of Pharmacy, Suzhou Science and Technology Town Hospital, Suzhou, China
| | - Xuechu Zhen
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| |
Collapse
|
59
|
Moretti F, Bergman P, Dodgson S, Marcellin D, Claerr I, Goodwin JM, DeJesus R, Kang Z, Antczak C, Begue D, Bonenfant D, Graff A, Genoud C, Reece-Hoyes JS, Russ C, Yang Z, Hoffman GR, Mueller M, Murphy LO, Xavier RJ, Nyfeler B. TMEM41B is a novel regulator of autophagy and lipid mobilization. EMBO Rep 2018; 19:e45889. [PMID: 30126924 PMCID: PMC6123663 DOI: 10.15252/embr.201845889] [Citation(s) in RCA: 123] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Revised: 07/03/2018] [Accepted: 07/12/2018] [Indexed: 01/22/2023] Open
Abstract
Autophagy maintains cellular homeostasis by targeting damaged organelles, pathogens, or misfolded protein aggregates for lysosomal degradation. The autophagic process is initiated by the formation of autophagosomes, which can selectively enclose cargo via autophagy cargo receptors. A machinery of well-characterized autophagy-related proteins orchestrates the biogenesis of autophagosomes; however, the origin of the required membranes is incompletely understood. Here, we have applied sensitized pooled CRISPR screens and identify the uncharacterized transmembrane protein TMEM41B as a novel regulator of autophagy. In the absence of TMEM41B, autophagosome biogenesis is stalled, LC3 accumulates at WIPI2- and DFCP1-positive isolation membranes, and lysosomal flux of autophagy cargo receptors and intracellular bacteria is impaired. In addition to defective autophagy, TMEM41B knockout cells display significantly enlarged lipid droplets and reduced mobilization and β-oxidation of fatty acids. Immunostaining and interaction proteomics data suggest that TMEM41B localizes to the endoplasmic reticulum (ER). Taken together, we propose that TMEM41B is a novel ER-localized regulator of autophagosome biogenesis and lipid mobilization.
Collapse
Affiliation(s)
| | - Phil Bergman
- Novartis Institutes for BioMedical Research, Cambridge, MA, USA
| | - Stacie Dodgson
- Harvard Medical School, Massachusetts General Hospital, Boston, MA, USA
| | - David Marcellin
- Novartis Institutes for BioMedical Research, Basel, Switzerland
| | - Isabelle Claerr
- Novartis Institutes for BioMedical Research, Basel, Switzerland
| | | | - Rowena DeJesus
- Novartis Institutes for BioMedical Research, Cambridge, MA, USA
| | - Zhao Kang
- Novartis Institutes for BioMedical Research, Cambridge, MA, USA
| | | | - Damien Begue
- Novartis Institutes for BioMedical Research, Basel, Switzerland
| | | | - Alexandra Graff
- Friedrich Miescher Institute for Biomedical Research, Basel, Switzerland
| | - Christel Genoud
- Friedrich Miescher Institute for Biomedical Research, Basel, Switzerland
| | | | - Carsten Russ
- Novartis Institutes for BioMedical Research, Cambridge, MA, USA
| | - Zinger Yang
- Novartis Institutes for BioMedical Research, Cambridge, MA, USA
| | | | | | - Leon O Murphy
- Novartis Institutes for BioMedical Research, Cambridge, MA, USA
| | - Ramnik J Xavier
- Harvard Medical School, Massachusetts General Hospital, Boston, MA, USA
| | - Beat Nyfeler
- Novartis Institutes for BioMedical Research, Basel, Switzerland
| |
Collapse
|
60
|
Cerveró C, Blasco A, Tarabal O, Casanovas A, Piedrafita L, Navarro X, Esquerda JE, Calderó J. Glial Activation and Central Synapse Loss, but Not Motoneuron Degeneration, Are Prevented by the Sigma-1 Receptor Agonist PRE-084 in the Smn2B/- Mouse Model of Spinal Muscular Atrophy. J Neuropathol Exp Neurol 2018; 77:577-597. [PMID: 29767748 DOI: 10.1093/jnen/nly033] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Spinal muscular atrophy (SMA) is characterized by the loss of α-motoneurons (MNs) with concomitant muscle denervation. MN excitability and vulnerability to disease are particularly regulated by cholinergic synaptic afferents (C-boutons), in which Sigma-1 receptor (Sig1R) is concentrated. Alterations in Sig1R have been associated with MN degeneration. Here, we investigated whether a chronic treatment with the Sig1R agonist PRE-084 was able to exert beneficial effects on SMA. We used a model of intermediate SMA, the Smn2B/- mouse, in which we performed a detailed characterization of the histopathological changes that occur throughout the disease. We report that Smn2B/- mice exhibited qualitative differences in major alterations found in mouse models of severe SMA: Smn2B/- animals showed more prominent MN degeneration, early motor axon alterations, marked changes in sensory neurons, and later MN deafferentation that correlated with conspicuous reactive gliosis and altered neuroinflammatory M1/M2 microglial balance. PRE-084 attenuated reactive gliosis, mitigated M1/M2 imbalance, and prevented MN deafferentation in Smn2B/- mice. These effects were also observed in a severe SMA model, the SMNΔ7 mouse. However, the prevention of gliosis and MN deafferentation promoted by PRE-084 were not accompanied by any improvements in clinical outcome or other major pathological changes found in SMA mice.
Collapse
Affiliation(s)
- Clàudia Cerveró
- Unitat de Neurobiologia Cel·lular, Departament de Medicina Experimental, Facultat de Medicina, Universitat de Lleida and Institut de Recerca Biomèdica de Lleida (IRBLLEIDA), Lleida, Catalonia, Spain
| | - Alba Blasco
- Unitat de Neurobiologia Cel·lular, Departament de Medicina Experimental, Facultat de Medicina, Universitat de Lleida and Institut de Recerca Biomèdica de Lleida (IRBLLEIDA), Lleida, Catalonia, Spain
| | - Olga Tarabal
- Unitat de Neurobiologia Cel·lular, Departament de Medicina Experimental, Facultat de Medicina, Universitat de Lleida and Institut de Recerca Biomèdica de Lleida (IRBLLEIDA), Lleida, Catalonia, Spain
| | - Anna Casanovas
- Unitat de Neurobiologia Cel·lular, Departament de Medicina Experimental, Facultat de Medicina, Universitat de Lleida and Institut de Recerca Biomèdica de Lleida (IRBLLEIDA), Lleida, Catalonia, Spain
| | - Lídia Piedrafita
- Unitat de Neurobiologia Cel·lular, Departament de Medicina Experimental, Facultat de Medicina, Universitat de Lleida and Institut de Recerca Biomèdica de Lleida (IRBLLEIDA), Lleida, Catalonia, Spain
| | - Xavier Navarro
- Group of Neuroplasticity and Regeneration, Institute of Neurosciences and Department of Cell Biology, Physiology and Immunology, Universitat Autònoma de Barcelona and CIBERNED, Bellaterra, Catalonia, Spain
| | - Josep E Esquerda
- Unitat de Neurobiologia Cel·lular, Departament de Medicina Experimental, Facultat de Medicina, Universitat de Lleida and Institut de Recerca Biomèdica de Lleida (IRBLLEIDA), Lleida, Catalonia, Spain
| | - Jordi Calderó
- Unitat de Neurobiologia Cel·lular, Departament de Medicina Experimental, Facultat de Medicina, Universitat de Lleida and Institut de Recerca Biomèdica de Lleida (IRBLLEIDA), Lleida, Catalonia, Spain
| |
Collapse
|
61
|
Abstract
Lipid droplets (LDs) are ubiquitous fat storage organelles and play key roles in lipid metabolism and energy homeostasis; in addition, they contribute to protein storage, folding, and degradation. However, a role for LDs in the nervous system remains largely unexplored. We discuss evidence supporting an intimate functional connection between LDs and motor neuron disease (MND) pathophysiology, examining how LD functions in systemic energy homeostasis, in neuron-glia metabolic coupling, and in protein folding and clearance may affect or contribute to disease pathology. An integrated understanding of LD biology and neurodegeneration may open the way for new therapeutic interventions.
Collapse
Affiliation(s)
- Giuseppa Pennetta
- Euan MacDonald Centre for Motor Neuron Disease Research, University of Edinburgh, Edinburgh EH8 9XD, UK; Centre for Integrative Physiology, University of Edinburgh, Edinburgh EH8 9XD, UK.
| | - Michael A Welte
- Department of Biology, University of Rochester, Rochester, NY 14627, USA
| |
Collapse
|
62
|
Abstract
Amyotrophic lateral sclerosis (ALS) is a devastating, uniformly lethal degenerative disorder of motor neurons that overlaps clinically with frontotemporal dementia (FTD). Investigations of the 10% of ALS cases that are transmitted as dominant traits have revealed numerous gene mutations and variants that either cause these disorders or influence their clinical phenotype. The evolving understanding of the genetic architecture of ALS has illuminated broad themes in the molecular pathophysiology of both familial and sporadic ALS and FTD. These central themes encompass disturbances of protein homeostasis, alterations in the biology of RNA binding proteins, and defects in cytoskeletal dynamics, as well as numerous downstream pathophysiological events. Together, these findings from ALS genetics provide new insight into therapies that target genetically distinct subsets of ALS and FTD.
Collapse
Affiliation(s)
- Mehdi Ghasemi
- Department of Neurology, University of Massachusetts Medical School, Worcester, Massachusetts 01655
| | - Robert H Brown
- Department of Neurology, University of Massachusetts Medical School, Worcester, Massachusetts 01655
| |
Collapse
|
63
|
Lehmann S, Esch E, Hartmann P, Goswami A, Nikolin S, Weis J, Beyer C, Johann S. Expression profile of pattern recognition receptors in skeletal muscle of SOD1 (G93A) amyotrophic lateral sclerosis (ALS) mice and sporadic ALS patients. Neuropathol Appl Neurobiol 2018; 44:606-627. [PMID: 29575052 DOI: 10.1111/nan.12483] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2017] [Accepted: 02/20/2018] [Indexed: 12/14/2022]
Abstract
AIMS Amyotrophic lateral sclerosis (ALS) is characterized by degeneration of motoneurons and progressive muscle wasting. Inflammatory processes, mediated by non-neuronal cells, such as glial cells, are known to contribute to disease progression. Inflammasomes consist of pattern recognition receptors (PRRs), apoptosis-associated speck-like protein (ASC) and caspase 1 and are essential for interleukin (IL) processing and a rapid immune response after tissue damage. Recently, we described inflammasome activation in the spinal cord of ALS patients and in SOD1(G93A) ALS mice. Since pathological changes in the skeletal muscle are early events in ALS, we hypothesized that PRRs might be abnormally expressed in muscle fibre degeneration. METHODS Western blot analysis, real-time PCR and immunohistochemistry were performed with muscle tissue from presymptomatic and early-symptomatic male SOD1(G93A) mice and with muscle biopsies of control and sporadic ALS (sALS) patients. Analysed PRRs include nucleotide-binding oligomerization domain-like (NOD-like) receptor protein 1 (NLRP1), NLR protein 3 (NLRP3), NLR family CARD domain-containing 4 (NLRC4) and absent in melanoma 2. Additionally, expression levels of ASC, caspase 1, interleukin 1 beta (IL1β) and interleukin 18 (IL18) were evaluated. RESULTS Expression of PRRs and ASC was detected in murine and human tissue. The PRR NLRC4, caspase 1 and IL1β were significantly elevated in denervated muscle of SOD1(G93A) mice and sALS patients. Furthermore, levels of caspase 1 and IL1β were already increased in presymptomatic animals. CONCLUSION Our findings suggest that increased inflammasome activation may be involved in skeletal muscle pathology in ALS. Furthermore, elevated levels of NLRC4, caspase 1 and IL1β reflect early changes in the skeletal muscle and may contribute to the denervation process.
Collapse
Affiliation(s)
- S Lehmann
- Institute of Neuroanatomy, Medical Clinic RWTH Aachen University, Aachen, Germany.,Institute Molecular and Cellular Anatomy (MOCA), Medical Clinic RWTH Aachen University, Aachen, Germany
| | - E Esch
- Institute of Neuroanatomy, Medical Clinic RWTH Aachen University, Aachen, Germany
| | - P Hartmann
- Institute of Neuroanatomy, Medical Clinic RWTH Aachen University, Aachen, Germany
| | - A Goswami
- Institute of Neuropathology, Medical Clinic RWTH Aachen University, Aachen, Germany
| | - S Nikolin
- Institute of Neuropathology, Medical Clinic RWTH Aachen University, Aachen, Germany
| | - J Weis
- Institute of Neuropathology, Medical Clinic RWTH Aachen University, Aachen, Germany
| | - C Beyer
- Institute of Neuroanatomy, Medical Clinic RWTH Aachen University, Aachen, Germany.,JARA - Translational Brain Medicine, Aachen, Germany
| | - S Johann
- Institute of Neuroanatomy, Medical Clinic RWTH Aachen University, Aachen, Germany.,Institute of Anatomy II, Medical Faculty Heinrich-Heine-University, Düsseldorf, Germany
| |
Collapse
|
64
|
Clark BJ, Prough RA, Klinge CM. Mechanisms of Action of Dehydroepiandrosterone. VITAMINS AND HORMONES 2018; 108:29-73. [PMID: 30029731 DOI: 10.1016/bs.vh.2018.02.003] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Dehydroepiandrosterone (3β-hydroxy-5-androsten-17-one, DHEA) and its sulfated metabolite DHEA-S are the most abundant steroids in circulation and decline with age. Rodent studies have shown that DHEA has a wide variety of effects on liver, kidney, adipose, reproductive tissues, and central nervous system/neuronal function. The mechanisms by which DHEA and DHEA-S impart their physiological effects may be direct actions on plasma membrane receptors, including a DHEA-specific, G-protein-coupled receptor in endothelial cells; various neuroreceptors, e.g., aminobutyric-acid-type A, N-methyl-d-aspartate (NMDA), and sigma-1 (S1R) receptors; by binding steroid receptors: androgen and estrogen receptors (ARs, ERα, or ERβ); or by their metabolism to more potent sex steroid hormones, e.g., testosterone, dihydrotestosterone, and estradiol, which bind with higher affinity to ARs and ERs. DHEA inhibits voltage-gated T-type calcium channels. DHEA activates peroxisome proliferator-activated receptor (PPARα) and CAR by a mechanism apparently involving PP2A, a protein phosphatase dephosphorylating PPARα and CAR to activate their transcriptional activity. We review our recent study showing DHEA activated GPER1 (G-protein-coupled estrogen receptor 1) in HepG2 cells to stimulate miR-21 transcription. This chapter reviews some of the physiological, biochemical, and molecular mechanisms of DHEA and DHEA-S activity.
Collapse
Affiliation(s)
- Barbara J Clark
- Department of Biochemistry and Molecular Genetics, Center for Genetics and Molecular Medicine, University of Louisville School of Medicine, Louisville, KY, United States
| | - Russell A Prough
- Department of Biochemistry and Molecular Genetics, Center for Genetics and Molecular Medicine, University of Louisville School of Medicine, Louisville, KY, United States
| | - Carolyn M Klinge
- Department of Biochemistry and Molecular Genetics, Center for Genetics and Molecular Medicine, University of Louisville School of Medicine, Louisville, KY, United States.
| |
Collapse
|
65
|
Medinas DB, Valenzuela V, Hetz C. Proteostasis disturbance in amyotrophic lateral sclerosis. Hum Mol Genet 2018; 26:R91-R104. [PMID: 28977445 DOI: 10.1093/hmg/ddx274] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2017] [Accepted: 07/11/2017] [Indexed: 12/11/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a progressive neurodegenerative disease affecting motoneurons in the brain and spinal cord leading to paralysis and death. Although the etiology of ALS remains poorly understood, abnormal protein aggregation and altered proteostasis are common features of sporadic and familial ALS forms. The proteostasis network is decomposed into different modules highly conserved across species and comprehends a collection of mechanisms related to protein synthesis, folding, trafficking, secretion and degradation that is distributed in different compartments inside the cell. Functional studies in various ALS models are revealing a complex scenario where distinct and even opposite effects in disease progression are observed depending on the targeted component of the proteostasis network. Importantly, alteration of the folding capacity of the endoplasmic reticulum (ER) is becoming a common pathological alteration in ALS, representing one of the earliest defects observed in disease models, contributing to denervation and motoneuron dysfunction. Strategies to target-specific components of the proteostasis network using small molecules and gene therapy are under development, and promise interesting avenues for future interventions to delay or stop ALS progression.
Collapse
Affiliation(s)
- Danilo B Medinas
- Biomedical Neuroscience Institute, Faculty of Medicine, University of Chile, Santiago, Chile.,Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago, Chile.,Center for Geroscience, Brain Health and Metabolism, Santiago, Chile
| | - Vicente Valenzuela
- Biomedical Neuroscience Institute, Faculty of Medicine, University of Chile, Santiago, Chile.,Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago, Chile.,Center for Geroscience, Brain Health and Metabolism, Santiago, Chile
| | - Claudio Hetz
- Biomedical Neuroscience Institute, Faculty of Medicine, University of Chile, Santiago, Chile.,Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago, Chile.,Center for Geroscience, Brain Health and Metabolism, Santiago, Chile.,Buck Institute for Research on Aging, Novato, CA, USA.,Department of Immunology and Infectious Diseases, Harvard School of Public Health, Boston, MA, USA
| |
Collapse
|
66
|
Lau DHW, Hartopp N, Welsh NJ, Mueller S, Glennon EB, Mórotz GM, Annibali A, Gomez-Suaga P, Stoica R, Paillusson S, Miller CCJ. Disruption of ER-mitochondria signalling in fronto-temporal dementia and related amyotrophic lateral sclerosis. Cell Death Dis 2018; 9:327. [PMID: 29491392 PMCID: PMC5832427 DOI: 10.1038/s41419-017-0022-7] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Revised: 09/18/2017] [Accepted: 09/20/2017] [Indexed: 12/13/2022]
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.
Collapse
Affiliation(s)
- Dawn H W Lau
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, 125 Coldharbour Lane Camberwell, London, SE5 9NU, UK
| | - Naomi Hartopp
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, 125 Coldharbour Lane Camberwell, London, SE5 9NU, UK
| | - Natalie J Welsh
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, 125 Coldharbour Lane Camberwell, London, SE5 9NU, UK
| | - Sarah Mueller
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, 125 Coldharbour Lane Camberwell, London, SE5 9NU, UK
| | - Elizabeth B Glennon
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, 125 Coldharbour Lane Camberwell, London, SE5 9NU, UK
| | - Gábor M Mórotz
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, 125 Coldharbour Lane Camberwell, London, SE5 9NU, UK
| | - Ambra Annibali
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, 125 Coldharbour Lane Camberwell, London, SE5 9NU, UK
| | - Patricia Gomez-Suaga
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, 125 Coldharbour Lane Camberwell, London, SE5 9NU, UK
| | - Radu Stoica
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, 125 Coldharbour Lane Camberwell, London, SE5 9NU, UK
| | - Sebastien Paillusson
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, 125 Coldharbour Lane Camberwell, London, SE5 9NU, UK.
| | - Christopher C J Miller
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, 125 Coldharbour Lane Camberwell, London, SE5 9NU, UK.
| |
Collapse
|
67
|
Endoplasmic reticulum and mitochondria in diseases of motor and sensory neurons: a broken relationship? Cell Death Dis 2018; 9:333. [PMID: 29491369 PMCID: PMC5832431 DOI: 10.1038/s41419-017-0125-1] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2017] [Revised: 09/25/2017] [Accepted: 10/10/2017] [Indexed: 12/13/2022]
Abstract
Recent progress in the understanding of neurodegenerative diseases revealed that multiple molecular mechanisms contribute to pathological changes in neurons. A large fraction of these alterations can be linked to dysfunction in the endoplasmic reticulum (ER) and mitochondria, affecting metabolism and secretion of lipids and proteins, calcium homeostasis, and energy production. Remarkably, these organelles are interacting with each other at specialized domains on the ER called mitochondria-associated membranes (MAMs). These membrane structures rely on the interaction of several complexes of proteins localized either at the mitochondria or at the ER interface and serve as an exchange platform of calcium, metabolites, and lipids, which are critical for the function of both organelles. In addition, recent evidence indicates that MAMs also play a role in the control of mitochondria dynamics and autophagy. MAMs thus start to emerge as a key element connecting many changes observed in neurodegenerative diseases. This review will focus on the role of MAMs in amyotrophic lateral sclerosis (ALS) and hereditary motor and sensory neuropathy, two neurodegenerative diseases particularly affecting neurons with long projecting axons. We will discuss how defects in MAM signaling may impair neuronal calcium homeostasis, mitochondrial dynamics, ER function, and autophagy, leading eventually to axonal degeneration. The possible impact of MAM dysfunction in glial cells, which may affect the capacity to support neurons and/or axons, will also be described. Finally, the possible role of MAMs as an interesting target for development of therapeutic interventions aiming at delaying or preventing neurodegeneration will be highlighted.
Collapse
|
68
|
Maurice T, Strehaiano M, Duhr F, Chevallier N. Amyloid toxicity is enhanced after pharmacological or genetic invalidation of the σ 1 receptor. Behav Brain Res 2018; 339:1-10. [PMID: 29129596 DOI: 10.1016/j.bbr.2017.11.010] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2017] [Accepted: 11/07/2017] [Indexed: 10/18/2022]
Abstract
The sigma-1 receptor (S1R) is a molecular chaperone which activity modulates several intracellular signals including calcium mobilization at mitochondria-associated endoplasmic reticulum membranes. S1R agonists are potent neuroprotectants against neurodegenerative insults and particularly in rodent models of Alzheimer's disease (AD). We here analyzed whether S1R inactivation modifies vulnerability to amyloid toxicity in AD models. Two strategies were used: (1) amyloid β[25-35] (Aβ25-35) peptide (1, 3, 9nmol) was injected intracerebroventricularly in mice treated repeatedly with the S1R antagonist NE-100 or in S1RKO mice, and (2) WT, APPSweInd, S1RKO, and APPSweInd/S1RKO mice were created and female littermates analyzed at 8 months of age. Learning deficits, oxidative stress, Bax level and BDNF content in the hippocampus were analyzed. Aβ25-35 induced learning impairment, oxidative stress, Bax induction and BDNF alteration at lower dose in NE-100-treated mice or S1RKO mice as compared to WT animals. The extent of learning deficits and biochemical alterations were also higher in APPSweInd/S1RKO mice as compared to WT, APPSweInd, and S1RKO animals. S1R inactivation or altered S1R expression augmented the pathological status in pharmacologic and genetic AD mouse models. These observations, in relation with the well-known protective effects of S1R agonists, are coherent with a role of signal amplifier in neurodegeneration and neuroprotection proposed for S1R in AD and related neurodegenerative disorders.
Collapse
Affiliation(s)
- Tangui Maurice
- MMDN, Univ. Montpellier, EPHE, INSERM, UMR-S1198, Montpellier, F-34095, France.
| | - Manon Strehaiano
- MMDN, Univ. Montpellier, EPHE, INSERM, UMR-S1198, Montpellier, F-34095, France
| | - Fanny Duhr
- MMDN, Univ. Montpellier, EPHE, INSERM, UMR-S1198, Montpellier, F-34095, France
| | - Nathalie Chevallier
- MMDN, Univ. Montpellier, EPHE, INSERM, UMR-S1198, Montpellier, F-34095, France
| |
Collapse
|
69
|
Impaired DNA damage response signaling by FUS-NLS mutations leads to neurodegeneration and FUS aggregate formation. Nat Commun 2018; 9:335. [PMID: 29362359 PMCID: PMC5780468 DOI: 10.1038/s41467-017-02299-1] [Citation(s) in RCA: 206] [Impact Index Per Article: 29.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2016] [Accepted: 11/20/2017] [Indexed: 02/08/2023] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is the most frequent motor neuron disease. Cytoplasmic fused in sarcoma (FUS) aggregates are pathological hallmarks of FUS-ALS. Proper shuttling between the nucleus and cytoplasm is essential for physiological cell function. However, the initial event in the pathophysiology of FUS-ALS remains enigmatic. Using human induced pluripotent stem cell (hiPSCs)-derived motor neurons (MNs), we show that impairment of poly(ADP-ribose) polymerase (PARP)-dependent DNA damage response (DDR) signaling due to mutations in the FUS nuclear localization sequence (NLS) induces additional cytoplasmic FUS mislocalization which in turn results in neurodegeneration and FUS aggregate formation. Our work suggests that a key pathophysiologic event in ALS is upstream of aggregate formation. Targeting DDR signaling could lead to novel therapeutic routes for ameliorating ALS. Abnormal cytoplasmic aggregates of FUS are a hallmark of some forms of amyotrophic lateral sclerosis (ALS). Here, using neurons derived from patients with FUS-ALS, the authors demonstrate that impairment of PARP-dependent DNA damage signaling is an event that occurs upstream of neurodegeneration and cytoplasmic aggregate formation in FUS-ALS.
Collapse
|
70
|
Maurel C, Dangoumau A, Marouillat S, Brulard C, Chami A, Hergesheimer R, Corcia P, Blasco H, Andres CR, Vourc'h P. Causative Genes in Amyotrophic Lateral Sclerosis and Protein Degradation Pathways: a Link to Neurodegeneration. Mol Neurobiol 2018; 55:6480-6499. [PMID: 29322304 DOI: 10.1007/s12035-017-0856-0] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Accepted: 12/20/2017] [Indexed: 12/11/2022]
Abstract
Amyotrophic lateral sclerosis (ALS) is a disease caused by the degeneration of motor neurons (MNs) leading to progressive muscle weakness and atrophy. Several molecular pathways have been implicated, such as glutamate-mediated excitotoxicity, defects in cytoskeletal dynamics and axonal transport, disruption of RNA metabolism, and impairments in proteostasis. ALS is associated with protein accumulation in the cytoplasm of cells undergoing neurodegeneration, which is a hallmark of the disease. In this review, we focus on mechanisms of proteostasis, particularly protein degradation, and discuss how they are related to the genetics of ALS. Indeed, the genetic bases of the disease with the implication of more than 30 genes associated with familial ALS to date, together with the important increase in understanding of endoplasmic reticulum (ER) stress, proteasomal degradation, and autophagy, allow researchers to better understand the mechanisms underlying the selective death of motor neurons in ALS. It is clear that defects in proteostasis are involved in this type of cellular degeneration, but whether or not these mechanisms are primary causes or merely consequential remains to be clearly demonstrated. Novel cellular and animal models allowing chronic expression of mutant proteins, for example, are required. Further studies linking genetic discoveries in ALS to mechanisms of protein clearance will certainly be crucial in order to accelerate translational and clinical research towards new therapeutic targets and strategies.
Collapse
Affiliation(s)
- C Maurel
- UMR INSERM U1253, Université de Tours, 37032, Tours, France
| | - A Dangoumau
- UMR INSERM U1253, Université de Tours, 37032, Tours, France
| | - S Marouillat
- UMR INSERM U1253, Université de Tours, 37032, Tours, France
| | - C Brulard
- UMR INSERM U1253, Université de Tours, 37032, Tours, France
| | - A Chami
- UMR INSERM U1253, Université de Tours, 37032, Tours, France
| | - R Hergesheimer
- UMR INSERM U1253, Université de Tours, 37032, Tours, France
| | - P Corcia
- UMR INSERM U1253, Université de Tours, 37032, Tours, France
- Service de Neurologie, CHRU de Tours, 37044, Tours, France
| | - H Blasco
- UMR INSERM U1253, Université de Tours, 37032, Tours, France
- Service de Biochimie et de Biologie Moléculaire, CHRU de Tours, 37044, Tours, France
| | - C R Andres
- UMR INSERM U1253, Université de Tours, 37032, Tours, France
- Service de Biochimie et de Biologie Moléculaire, CHRU de Tours, 37044, Tours, France
| | - P Vourc'h
- UMR INSERM U1253, Université de Tours, 37032, Tours, France.
- Service de Biochimie et de Biologie Moléculaire, CHRU de Tours, 37044, Tours, France.
| |
Collapse
|
71
|
Penke B, Fülöp L, Szűcs M, Frecska E. The Role of Sigma-1 Receptor, an Intracellular Chaperone in Neurodegenerative Diseases. Curr Neuropharmacol 2018; 16:97-116. [PMID: 28554311 PMCID: PMC5771390 DOI: 10.2174/1570159x15666170529104323] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Revised: 04/15/2017] [Accepted: 05/25/2017] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Widespread protein aggregation occurs in the living system under stress or during aging, owing to disturbance of endoplasmic reticulum (ER) proteostasis. Many neurodegenerative diseases may have a common mechanism: the failure of protein homeostasis. Perturbation of ER results in unfolded protein response (UPR). Prolonged chronical UPR may activate apoptotic pathways and cause cell death. METHODS Research articles on Sigma-1 receptor were reviewed. RESULTS ER is associated to mitochondria by the mitochondria-associated ER-membrane, MAM. The sigma-1 receptor (Sig-1R), a well-known ER-chaperone localizes in the MAM. It serves for Ca2+-signaling between the ER and mitochondria, involved in ion channel activities and especially important during neuronal differentiation. Sig-1R acts as central modulator in inter-organelle signaling. Sig-1R helps cell survival by attenuating ER-stress. According to sequence based predictions Sig-1R is a 223 amino acid protein with two transmembrane (2TM) domains. The X-ray structure of the Sig-1R [1] showed a membrane-bound trimeric assembly with one transmembrane (1TM) region. Despite the in vitro determined assembly, the results of in vivo studies are rather consistent with the 2TM structure. The receptor has unique and versatile pharmacological profile. Dimethyl tryptamine (DMT) and neuroactive steroids are endogenous ligands that activate Sig-1R. The receptor has a plethora of interacting client proteins. Sig-1R exists in oligomeric structures (dimer-trimer-octamer-multimer) and this fact may explain interaction with diverse proteins. CONCLUSION Sig-1R agonists have been used in the treatment of different neurodegenerative diseases, e.g. Alzheimer's and Parkinson's diseases (AD and PD) and amyotrophic lateral sclerosis. Utilization of Sig-1R agents early in AD and similar other diseases has remained an overlooked therapeutic opportunity.
Collapse
Affiliation(s)
- Botond Penke
- University of Szeged, Department of Medical Chemistry, Faculty of Medicine, Szeged, Hungary
| | - Lívia Fülöp
- University of Szeged, Department of Medical Chemistry, Faculty of Medicine, Szeged, Hungary
| | - Mária Szűcs
- University of Szeged, Department of Medical Chemistry, Faculty of Medicine, Szeged, Hungary
| | - Ede Frecska
- University of Debrecen, Department of Psychiatry, Faculty of Medicine, Debrecen, Hungary
| |
Collapse
|
72
|
Hong J, Wang L, Zhang T, Zhang B, Chen L. Sigma-1 receptor knockout increases α-synuclein aggregation and phosphorylation with loss of dopaminergic neurons in substantia nigra. Neurobiol Aging 2017; 59:171-183. [PMID: 28870519 DOI: 10.1016/j.neurobiolaging.2017.08.007] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2017] [Revised: 06/20/2017] [Accepted: 08/02/2017] [Indexed: 01/17/2023]
Abstract
Sigma-1 receptor (σ1R) is expressed in dopaminergic neurons of substantia nigra. Here, we show that σ1R knockout (σ1R-/-) mice, at age 6-12 months, appeared with age-related loss of dopaminergic neurons and decline of motor coordination. Levels of α-synuclein (αSyn) oligomers and fibrillar αSyn in substantia nigra of σ1R-/- mice were age-dependently increased without the changes in αSyn monomers. The phosphorylation of αSyn monomers or oligomers in dopaminergic neurons was enhanced in σ1R-/- mice. Levels of phosphorylated eIF2a and C/EBP homologous protein expression were elevated in σ1R-/- mice with decline of proteasome activity. Inhibition of endoplasmic reticulum stress by salubrinal recovered the αSyn phosphorylation and proteasome activity and prevented early oligomerization of αSyn in σ1R-/- mice. Rifampicin reduced the late increase of αSyn oligomers in σ1R-/- mice. Rifampicin or salubrinal could reduce the loss of dopaminergic neurons in σ1R-/- mice and improved their motor coordination. The results indicate that the σ1R deficiency through enhanced aggregation and phosphorylation of αSyn causes the loss of dopaminergic neurons leading to the decline of motor coordination.
Collapse
Affiliation(s)
- Juan Hong
- State Key Lab of Reproductive Medicine, Nanjing Medical University, Nanjing, China; Department of Physiology, Nanjing Medical University, Nanjing, China
| | - Ling Wang
- Department of Physiology, Nanjing Medical University, Nanjing, China
| | - Tingting Zhang
- Department of Physiology, Nanjing Medical University, Nanjing, China
| | - Baofeng Zhang
- Department of Physiology, Nanjing Medical University, Nanjing, China
| | - Ling Chen
- State Key Lab of Reproductive Medicine, Nanjing Medical University, Nanjing, China; Department of Physiology, Nanjing Medical University, Nanjing, China.
| |
Collapse
|
73
|
Pérez MJ, Ponce DP, Osorio-Fuentealba C, Behrens MI, Quintanilla RA. Mitochondrial Bioenergetics Is Altered in Fibroblasts from Patients with Sporadic Alzheimer's Disease. Front Neurosci 2017; 11:553. [PMID: 29056898 PMCID: PMC5635042 DOI: 10.3389/fnins.2017.00553] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Accepted: 09/20/2017] [Indexed: 12/29/2022] Open
Abstract
The identification of an early biomarker to diagnose Alzheimer's disease (AD) remains a challenge. Neuropathological studies in animal and AD patients have shown that mitochondrial dysfunction is a hallmark of the development of the disease. Current studies suggest the use of peripheral tissues, like skin fibroblasts as a possibility to detect the early pathological alterations present in the AD brain. In this context, we studied mitochondrial function properties (bioenergetics and morphology) in cultured fibroblasts obtained from AD, aged-match and young healthy patients. We observed that AD fibroblasts presented a significant reduction in mitochondrial length with important changes in the expression of proteins that control mitochondrial fusion. Moreover, AD fibroblasts showed a distinct alteration in proteolytic processing of OPA1, a master regulator of mitochondrial fusion, compared to control fibroblasts. Complementary to these changes AD fibroblasts showed a dysfunctional mitochondrial bioenergetics profile that differentiates these cells from aged-matched and young patient fibroblasts. Our findings suggest that the human skin fibroblasts obtained from AD patients could replicate mitochondrial impairment observed in the AD brain. These promising observations suggest that the analysis of mitochondrial bioenergetics could represent a promising strategy to develop new diagnostic methods in peripheral tissues of AD patients.
Collapse
Affiliation(s)
- María J Pérez
- Laboratory of Neurodegenerative Diseases, Universidad Autónoma de Chile, Santiago, Chile.,Centro de Investigación y Estudio del Consumo de Alcohol en Adolescentes, Santiago, Chile
| | - Daniela P Ponce
- Instituto de Ciencias Biomédicas, Universidad de Chile, Santiago, Chile
| | - Cesar Osorio-Fuentealba
- Centro de Investigación y Estudio del Consumo de Alcohol en Adolescentes, Santiago, Chile.,Departamento Kinesiología, Universidad Metropolitana de Ciencias de la Educación, Ñuñoa, Chile
| | - Maria I Behrens
- Instituto de Ciencias Biomédicas, Universidad de Chile, Santiago, Chile
| | - Rodrigo A Quintanilla
- Laboratory of Neurodegenerative Diseases, Universidad Autónoma de Chile, Santiago, Chile.,Centro de Investigación y Estudio del Consumo de Alcohol en Adolescentes, Santiago, Chile
| |
Collapse
|
74
|
Ji LL, Peng JB, Fu CH, Tong L, Wang ZY. Sigma-1 receptor activation ameliorates anxiety-like behavior through NR2A-CREB-BDNF signaling pathway in a rat model submitted to single-prolonged stress. Mol Med Rep 2017; 16:4987-4993. [PMID: 28791385 DOI: 10.3892/mmr.2017.7185] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2017] [Accepted: 07/11/2017] [Indexed: 11/05/2022] Open
Abstract
Accumulating evidence has demonstrated that the σ-1 receptor (σ‑1R) possesses neuroprotective effects and is a potential novel therapeutic target for certain psychiatric diseases, including post‑traumatic stress disorder (PTSD) accompanied with anxiety disorder. It has been reported that σ‑1R agonist treatment could be modulated by the brain‑derived neurotrophic factor (BDNF) signaling pathway. However, it remains unclear whether BDNF and its upstream regulator are mechanistically involved in the therapeutic effect of σ‑1R in PTSD. To address this question, rats were subjected to a single‑prolonged stress (SPS) paradigm and σ‑1R agonist administration. Open‑field and elevated plus maze tests were implemented to evaluate the effect of σ‑1R activation on the improvement of anxiety‑like behaviors. Furthermore, the expression levels of BDNF, phosphorylated cAMP responsive element‑binding protein (CREB) and glutamate receptor ionotropic N‑methyl D‑aspartate 2A (NMDAR2A) were investigated in the hippocampi of rats. It was revealed that the downregulation of BDNF, phosphorylated CREB and NMDAR2A induced by SPS were reversed by σ‑1R activation. Collectively, the results of the present study suggest that the NMDAR2A/CREB/BDNF signaling pathway is involved in the activation of σ‑1R resulting in therapeutic effects for PTSD.
Collapse
Affiliation(s)
- Li-Li Ji
- Department of Anatomy, College of Basic Medical Sciences, China Medical University, Shenyang, Liaoning 110122, P.R. China
| | - Jun-Bo Peng
- Department of Anatomy, College of Basic Medical Sciences, China Medical University, Shenyang, Liaoning 110122, P.R. China
| | - Chang-Hai Fu
- Department of Anatomy, College of Basic Medical Sciences, China Medical University, Shenyang, Liaoning 110122, P.R. China
| | - Lei Tong
- Department of Anatomy, College of Basic Medical Sciences, China Medical University, Shenyang, Liaoning 110122, P.R. China
| | - Zhen-Yu Wang
- Department of Anatomy, College of Basic Medical Sciences, China Medical University, Shenyang, Liaoning 110122, P.R. China
| |
Collapse
|
75
|
Dreser A, Vollrath JT, Sechi A, Johann S, Roos A, Yamoah A, Katona I, Bohlega S, Wiemuth D, Tian Y, Schmidt A, Vervoorts J, Dohmen M, Beyer C, Anink J, Aronica E, Troost D, Weis J, Goswami A. The ALS-linked E102Q mutation in Sigma receptor-1 leads to ER stress-mediated defects in protein homeostasis and dysregulation of RNA-binding proteins. Cell Death Differ 2017; 24:1655-1671. [PMID: 28622300 PMCID: PMC5596426 DOI: 10.1038/cdd.2017.88] [Citation(s) in RCA: 80] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2015] [Revised: 03/14/2017] [Accepted: 03/22/2017] [Indexed: 12/21/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is characterized by the selective degeneration of motor neurons (MNs) and their target muscles. Misfolded proteins which often form intracellular aggregates are a pathological hallmark of ALS. Disruption of the functional interplay between protein degradation (ubiquitin proteasome system and autophagy) and RNA-binding protein homeostasis has recently been suggested as an integrated model that merges several ALS-associated proteins into a common pathophysiological pathway. The E102Q mutation in one such candidate gene, the endoplasmic reticulum (ER) chaperone Sigma receptor-1 (SigR1), has been reported to cause juvenile ALS. Although loss of SigR1 protein contributes to neurodegeneration in several ways, the molecular mechanisms underlying E102Q-SigR1-mediated neurodegeneration are still unclear. In the present study, we showed that the E102Q-SigR1 protein rapidly aggregates and accumulates in the ER and associated compartments in transfected cells, leading to structural alterations of the ER, nuclear envelope and mitochondria and to subsequent defects in proteasomal degradation and calcium homeostasis. ER defects and proteotoxic stress generated by E102Q-SigR1 aggregates further induce autophagy impairment, accumulation of stress granules and cytoplasmic aggregation of the ALS-linked RNA-binding proteins (RBPs) matrin-3, FUS, and TDP-43. Similar ultrastructural abnormalities as well as altered protein degradation and misregulated RBP homeostasis were observed in primary lymphoblastoid cells (PLCs) derived from E102Q-SigR1 fALS patients. Consistent with these findings, lumbar α-MNs of both sALS as well as fALS patients showed cytoplasmic matrin-3 aggregates which were not co-localized with pTDP-43 aggregates. Taken together, our results support the notion that E102Q-SigR1-mediated ALS pathogenesis comprises a synergistic mechanism of both toxic gain and loss of function involving a vicious circle of altered ER function, impaired protein homeostasis and defective RBPs.
Collapse
Affiliation(s)
- Alice Dreser
- Institute of Neuropathology, RWTH Aachen University Medical School, Aachen, Germany
| | - Jan Tilmann Vollrath
- Institute of Neuropathology, RWTH Aachen University Medical School, Aachen, Germany
| | - Antonio Sechi
- Institute of Biomedical Engineering, Deparment of Cell Biology, RWTH Aachen University Medical School, Aachen, Germany
| | - Sonja Johann
- Institute of Neuroanatomy, RWTH Aachen University Medical School, Aachen, Germany
| | - Andreas Roos
- Institute of Neuropathology, RWTH Aachen University Medical School, Aachen, Germany
- Leibniz-Institut für Analytische Wissenschaften – ISAS – e.V., Dortmund, Germany
- Institute of Genetic Medicine, John Walton Muscular Dystrophy Research Centre, International Centre for Life, Central Parkway, Newcastle upon Tyne, England, UK
| | - Alfred Yamoah
- Institute of Neuropathology, RWTH Aachen University Medical School, Aachen, Germany
| | - Istvan Katona
- Institute of Neuropathology, RWTH Aachen University Medical School, Aachen, Germany
| | - Saeed Bohlega
- Department of Genetics, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Dominik Wiemuth
- Institute of Physiology, RWTH Aachen University Medical School, Aachen Germany
| | - Yuemin Tian
- Institute of Physiology, RWTH Aachen University Medical School, Aachen Germany
| | - Axel Schmidt
- Institute of Physiology, RWTH Aachen University Medical School, Aachen Germany
| | - Jörg Vervoorts
- Institute of Biochemistry and Molecular Biology, RWTH Aachen University Medical School, Aachen, Germany
| | - Marc Dohmen
- Institute of Biochemistry and Molecular Biology, RWTH Aachen University Medical School, Aachen, Germany
| | - Cordian Beyer
- Institute of Neuroanatomy, RWTH Aachen University Medical School, Aachen, Germany
| | - Jasper Anink
- Department of (Neuro)Pathology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Eleonora Aronica
- Department of (Neuro)Pathology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Dirk Troost
- Department of (Neuro)Pathology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Joachim Weis
- Institute of Neuropathology, RWTH Aachen University Medical School, Aachen, Germany
| | - Anand Goswami
- Institute of Neuropathology, RWTH Aachen University Medical School, Aachen, Germany
| |
Collapse
|
76
|
Tadić V, Malci A, Goldhammer N, Stubendorff B, Sengupta S, Prell T, Keiner S, Liu J, Guenther M, Frahm C, Witte OW, Grosskreutz J. Sigma 1 receptor activation modifies intracellular calcium exchange in the G93A hSOD1 ALS model. Neuroscience 2017; 359:105-118. [PMID: 28723387 DOI: 10.1016/j.neuroscience.2017.07.012] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2016] [Revised: 06/30/2017] [Accepted: 07/07/2017] [Indexed: 10/19/2022]
Abstract
Aberrations in intracellular calcium (Ca2+) have been well established within amyotrophic lateral sclerosis (ALS), a severe motor neuron disease. Intracellular Ca2+ concentration is controlled in part through the endoplasmic reticulum (ER) mitochondria Ca2+ cycle (ERMCC). The ER supplies Ca2+ to the mitochondria at close contacts between the two organelles, i.e. the mitochondria-associated ER membranes (MAMs). The Sigma 1 receptor (Sig1R) is enriched at MAMs, where it acts as an inter-organelle signaling modulator. However, its impact on intracellular Ca2+ at the cellular level remains to be thoroughly investigated. Here, we used cultured embryonic mice spinal neurons to investigate the influence of Sig1R activation on intracellular Ca2+ homeostasis in the presence of G93AhSOD1 (G93A), an established ALS-causing mutation. Sig1R expression was increased in G93A motor neurons relative to non-transgenic (nontg) controls. Furthermore, we demonstrated significantly reduced bradykinin-sensitive intracellular Ca2+ stores in G93A spinal neurons, which were normalized by the Sig1R agonist SA4503. Moreover, SA4503 accelerated cytosolic Ca2+ clearance following a) AMPAR activation by kainate and b) IP3R-mediated ER Ca2+ release following bradykinin stimulation in both genotypes. PRE-084 (another Sig1R agonist) did not exert any significant effects on cytosolic Ca2+. Both Sig1R expression and functionality were altered by the G93A mutation, indicating the centrality of Sig1R in ALS pathology. Here, we showed that intracellular Ca2+ shuttling can be manipulated by Sig1R activation, thus demonstrating the value of using the pharmacological manipulation of Sig1R to understand Ca2+ homeostasis.
Collapse
Affiliation(s)
- Vedrana Tadić
- Hans Berger Department of Neurology, Jena University Hospital, Am Klinikum 1, 07747 Jena, Germany.
| | - Ayse Malci
- Hans Berger Department of Neurology, Jena University Hospital, Am Klinikum 1, 07747 Jena, Germany.
| | - Nadine Goldhammer
- Hans Berger Department of Neurology, Jena University Hospital, Am Klinikum 1, 07747 Jena, Germany.
| | - Beatrice Stubendorff
- Hans Berger Department of Neurology, Jena University Hospital, Am Klinikum 1, 07747 Jena, Germany.
| | - Saikata Sengupta
- Hans Berger Department of Neurology, Jena University Hospital, Am Klinikum 1, 07747 Jena, Germany.
| | - Tino Prell
- Hans Berger Department of Neurology, Jena University Hospital, Am Klinikum 1, 07747 Jena, Germany.
| | - Silke Keiner
- Hans Berger Department of Neurology, Jena University Hospital, Am Klinikum 1, 07747 Jena, Germany.
| | - Jingyu Liu
- Hans Berger Department of Neurology, Jena University Hospital, Am Klinikum 1, 07747 Jena, Germany.
| | - Madlen Guenther
- Hans Berger Department of Neurology, Jena University Hospital, Am Klinikum 1, 07747 Jena, Germany.
| | - Christiane Frahm
- Hans Berger Department of Neurology, Jena University Hospital, Am Klinikum 1, 07747 Jena, Germany.
| | - Otto W Witte
- Hans Berger Department of Neurology, Jena University Hospital, Am Klinikum 1, 07747 Jena, Germany.
| | - Julian Grosskreutz
- Hans Berger Department of Neurology, Jena University Hospital, Am Klinikum 1, 07747 Jena, Germany.
| |
Collapse
|
77
|
Weng TY, Tsai SYA, Su TP. Roles of sigma-1 receptors on mitochondrial functions relevant to neurodegenerative diseases. J Biomed Sci 2017; 24:74. [PMID: 28917260 PMCID: PMC5603014 DOI: 10.1186/s12929-017-0380-6] [Citation(s) in RCA: 80] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Accepted: 09/05/2017] [Indexed: 12/20/2022] Open
Abstract
The sigma-1 receptor (Sig-1R) is a chaperone that resides mainly at the mitochondrion-associated endoplasmic reticulum (ER) membrane (called the MAMs) and acts as a dynamic pluripotent modulator in living systems. At the MAM, the Sig-1R is known to play a role in regulating the Ca2+ signaling between ER and mitochondria and in maintaining the structural integrity of the MAM. The MAM serves as bridges between ER and mitochondria regulating multiple functions such as Ca2+ transfer, energy exchange, lipid synthesis and transports, and protein folding that are pivotal to cell survival and defense. Recently, emerging evidences indicate that the MAM is critical in maintaining neuronal homeostasis. Thus, given the specific localization of the Sig-1R at the MAM, we highlight and propose that the direct or indirect regulations of the Sig-1R on mitochondrial functions may relate to neurodegenerative diseases including Alzheimer's disease (AD), Parkinson's disease (PD), Huntington's disease (HD) and amyotrophic lateral sclerosis (ALS). In addition, the promising use of Sig-1R ligands to rescue mitochondrial dysfunction-induced neurodegeneration is addressed.
Collapse
Affiliation(s)
- Tzu-Yu Weng
- Cellular Pathobiology Section, Integrative Neuroscience Branch, Intramural Research Program, National Institute on Drug Abuse, NIH, DHHS, IRP, NIDA/NIH, Triad Bldg. suite 3512, 333 Cassell Drive, Baltimore, MD 21224 USA
- Genomics Research Center, Academia Sinica, Taipei, Taiwan
| | - Shang-Yi Anne Tsai
- Cellular Pathobiology Section, Integrative Neuroscience Branch, Intramural Research Program, National Institute on Drug Abuse, NIH, DHHS, IRP, NIDA/NIH, Triad Bldg. suite 3512, 333 Cassell Drive, Baltimore, MD 21224 USA
| | - Tsung-Ping Su
- Cellular Pathobiology Section, Integrative Neuroscience Branch, Intramural Research Program, National Institute on Drug Abuse, NIH, DHHS, IRP, NIDA/NIH, Triad Bldg. suite 3512, 333 Cassell Drive, Baltimore, MD 21224 USA
| |
Collapse
|
78
|
Wang J, Saul A, Cui X, Roon P, Smith SB. Absence of Sigma 1 Receptor Accelerates Photoreceptor Cell Death in a Murine Model of Retinitis Pigmentosa. Invest Ophthalmol Vis Sci 2017; 58:4545-4558. [PMID: 28877319 PMCID: PMC5586962 DOI: 10.1167/iovs.17-21947] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2017] [Accepted: 07/31/2017] [Indexed: 02/06/2023] Open
Abstract
Purpose Sigma 1 Receptor (Sig1R) is a novel therapeutic target in neurodegenerative diseases, including retinal disease. Sig1R-/- mice have late-onset retinal degeneration with ganglion cell loss that worsens under stress. Whether Sig1R plays a role in maintaining other retinal neurons is unknown, but was investigated here using rd10 mice, a model of severe photoreceptor degeneration. Methods Wild-type, rd10, and rd10/Sig1R-/- mice were subjected to ERG and spectral-domain optical coherence tomography (SD-OCT) to assess visual function/structure in situ. Retinas imaged microscopically were subjected to morphometric analysis, immunodetection of cones, and analysis of gliosis. Oxidative and endoplasmic reticulum (ER) stress was evaluated at mRNA/protein levels. Results Photopic ERG responses were reduced significantly in rd10/Sig1R-/- versus rd10 mice at P28 (31 ± 6 vs. 56 ± 7 μV), indicating accelerated cone loss when Sig1R was absent. At P28, SD-OCT revealed reduced retinal thickness in rd10/Sig1R-/- mice (60% of WT) versus rd10 (80% of WT). Morphometric analysis disclosed profound photoreceptor nuclei loss in rd10/Sig1R-/- versus rd10 mice. rd10/Sig1R-/- mice had 35% and 60% fewer photoreceptors, respectively, at P28 and P35, than rd10. Peanut agglutinin cone labeling decreased significantly; gliosis increased significantly in rd10/Sig1R-/- versus rd10 mice. At P21, NRF2 levels increased in rd10/Sig1R-/- mice versus rd10 and downstream antioxidants increased indicating oxidative stress. At P28, ER stress genes/proteins, especially XBP1, a potent transcriptional activator of the unfolded protein response and CHOP, a proapoptotic transcription factor, increased significantly in rd10/Sig1R-/- mice versus rd10. Conclusions Photoreceptor cell degeneration accelerates and cone function diminishes much earlier in rd10/Sig1R-/- than rd10 mice emphasizing the importance of Sig1R as a modulator of retinal cell survival.
Collapse
Affiliation(s)
- Jing Wang
- Department of Cellular Biology and Anatomy, The Medical College of Georgia at Augusta University, Augusta, Georgia, United States
- The James and Jean Culver Vision Discovery Institute, Augusta University, Augusta, Georgia, United States
| | - Alan Saul
- The James and Jean Culver Vision Discovery Institute, Augusta University, Augusta, Georgia, United States
- Department of Ophthalmology, Augusta University, Augusta, Georgia, United States
| | - Xuezhi Cui
- Department of Cellular Biology and Anatomy, The Medical College of Georgia at Augusta University, Augusta, Georgia, United States
- The James and Jean Culver Vision Discovery Institute, Augusta University, Augusta, Georgia, United States
| | - Penny Roon
- Department of Cellular Biology and Anatomy, The Medical College of Georgia at Augusta University, Augusta, Georgia, United States
| | - Sylvia B. Smith
- Department of Cellular Biology and Anatomy, The Medical College of Georgia at Augusta University, Augusta, Georgia, United States
- The James and Jean Culver Vision Discovery Institute, Augusta University, Augusta, Georgia, United States
- Department of Ophthalmology, Augusta University, Augusta, Georgia, United States
| |
Collapse
|
79
|
Kollipara L, Buchkremer S, Coraspe JAG, Hathazi D, Senderek J, Weis J, Zahedi RP, Roos A. In-depth phenotyping of lymphoblastoid cells suggests selective cellular vulnerability in Marinesco-Sjögren syndrome. Oncotarget 2017; 8:68493-68516. [PMID: 28978133 PMCID: PMC5620273 DOI: 10.18632/oncotarget.19663] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2016] [Accepted: 05/28/2017] [Indexed: 12/18/2022] Open
Abstract
SIL1 is a ubiquitous protein of the Endoplasmic Reticulum (ER) acting as a co-chaperone for the ER-resident chaperone, BiP. Recessive mutations of the corresponding gene lead to vulnerability of skeletal muscle and central nervous system in man (Marinesco-Sjögren syndrome; MSS) and mouse. However, it is still unclear how loss of ubiquitous SIL1 leads to selective vulnerability of the nervous system and skeletal muscle whereas other cells and organs are protected from clinical manifestations. In this study we aimed to disentangle proteins participating in selective vulnerability of SIL1-deficient cells and tissues: morphological examination of MSS patient-derived lymphoblastoid cells revealed altered organelle structures (ER, nucleus and mitochondria) thus showing subclinical vulnerability. To correlate structural perturbations with biochemical changes and to identify proteins potentially preventing phenotypical manifestation, proteomic studies have been carried out. Results of proteomic profiling are in line with the morphological findings and show affection of nuclear, mitochondrial and cytoskeletal proteins as well as of such responsible for cellular viability. Moreover, expression patterns of proteins known to be involved in neuromuscular disorders or in development and function of the nervous system were altered. Paradigmatic findings were confirmed by immunohistochemistry of splenic lymphocytes and the cerebellum of SIL1-deficient mice. Ataxin-10, identified with increased abundance in our proteome profile, is necessary for the neuronal survival but also controls muscle fiber apoptosis, thus declaring this protein as a plausible candidate for selective tissue vulnerability. Our combined results provide first insights into the molecular causes of selective cell and tissue vulnerability defining the MSS phenotype.
Collapse
Affiliation(s)
- Laxmikanth Kollipara
- Leibniz-Institut für Analytische Wissenschaften-ISAS -e.V., 44227 Dortmund, Germany
| | - Stephan Buchkremer
- Institute of Neuropathology, University Hospital Aachen, RWTH Aachen, 5274 Aachen, Germany
| | | | - Denisa Hathazi
- Leibniz-Institut für Analytische Wissenschaften-ISAS -e.V., 44227 Dortmund, Germany
| | - Jan Senderek
- Friedrich-Baur-Institute, Medical Faculty, Ludwig-Maximilians-University, 80336 Munich, Germany
| | - Joachim Weis
- Institute of Neuropathology, University Hospital Aachen, RWTH Aachen, 5274 Aachen, Germany
| | - René P Zahedi
- Leibniz-Institut für Analytische Wissenschaften-ISAS -e.V., 44227 Dortmund, Germany
| | - Andreas Roos
- Leibniz-Institut für Analytische Wissenschaften-ISAS -e.V., 44227 Dortmund, Germany.,Institute of Neuropathology, University Hospital Aachen, RWTH Aachen, 5274 Aachen, Germany.,The John Walton Muscular Dystrophy Research Centre, MRC Centre for Neuromuscular Diseases, Newcastle University, Newcastle upon Tyne, NE1 3BZ, UK
| |
Collapse
|
80
|
Szymański J, Janikiewicz J, Michalska B, Patalas-Krawczyk P, Perrone M, Ziółkowski W, Duszyński J, Pinton P, Dobrzyń A, Więckowski MR. Interaction of Mitochondria with the Endoplasmic Reticulum and Plasma Membrane in Calcium Homeostasis, Lipid Trafficking and Mitochondrial Structure. Int J Mol Sci 2017; 18:ijms18071576. [PMID: 28726733 PMCID: PMC5536064 DOI: 10.3390/ijms18071576] [Citation(s) in RCA: 184] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2017] [Revised: 07/10/2017] [Accepted: 07/13/2017] [Indexed: 12/12/2022] Open
Abstract
Studying organelles in isolation has been proven to be indispensable for deciphering the underlying mechanisms of molecular cell biology. However, observing organelles in intact cells with the use of microscopic techniques reveals a new set of different junctions and contact sites between them that contribute to the control and regulation of various cellular processes, such as calcium and lipid exchange or structural reorganization of the mitochondrial network. In recent years, many studies focused their attention on the structure and function of contacts between mitochondria and other organelles. From these studies, findings emerged showing that these contacts are involved in various processes, such as lipid synthesis and trafficking, modulation of mitochondrial morphology, endoplasmic reticulum (ER) stress, apoptosis, autophagy, inflammation and Ca2+ handling. In this review, we focused on the physical interactions of mitochondria with the endoplasmic reticulum and plasma membrane and summarized present knowledge regarding the role of mitochondria-associated membranes in calcium homeostasis and lipid metabolism.
Collapse
Affiliation(s)
- Jędrzej Szymański
- Department of Biochemistry, Nencki Institute of Experimental Biology, Pasteur 3, 02-093 Warsaw, Poland.
| | - Justyna Janikiewicz
- Department of Biochemistry, Nencki Institute of Experimental Biology, Pasteur 3, 02-093 Warsaw, Poland.
| | - Bernadeta Michalska
- Department of Biochemistry, Nencki Institute of Experimental Biology, Pasteur 3, 02-093 Warsaw, Poland.
| | - Paulina Patalas-Krawczyk
- Department of Biochemistry, Nencki Institute of Experimental Biology, Pasteur 3, 02-093 Warsaw, Poland.
| | - Mariasole Perrone
- Department of Morphology, Surgery and Experimental Medicine, Section of Pathology, Oncology and Experimental Biology, Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, 44121 Ferrara, Italy.
| | - Wiesław Ziółkowski
- Department of Bioenergetics and Nutrition, Gdańsk University of Physical Education and Sport, 80-336 Gdańsk, Poland.
| | - Jerzy Duszyński
- Department of Biochemistry, Nencki Institute of Experimental Biology, Pasteur 3, 02-093 Warsaw, Poland.
| | - Paolo Pinton
- Department of Morphology, Surgery and Experimental Medicine, Section of Pathology, Oncology and Experimental Biology, Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, 44121 Ferrara, Italy.
| | - Agnieszka Dobrzyń
- Department of Biochemistry, Nencki Institute of Experimental Biology, Pasteur 3, 02-093 Warsaw, Poland.
| | - Mariusz R Więckowski
- Department of Biochemistry, Nencki Institute of Experimental Biology, Pasteur 3, 02-093 Warsaw, Poland.
| |
Collapse
|
81
|
Gueguinou M, Crottès D, Chantôme A, Rapetti-Mauss R, Potier-Cartereau M, Clarysse L, Girault A, Fourbon Y, Jézéquel P, Guérin-Charbonnel C, Fromont G, Martin P, Pellissier B, Schiappa R, Chamorey E, Mignen O, Uguen A, Borgese F, Vandier C, Soriani O. The SigmaR1 chaperone drives breast and colorectal cancer cell migration by tuning SK3-dependent Ca 2+ homeostasis. Oncogene 2017; 36:3640-3647. [PMID: 28114279 DOI: 10.1038/onc.2016.501] [Citation(s) in RCA: 91] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2016] [Revised: 11/19/2016] [Accepted: 11/28/2016] [Indexed: 12/21/2022]
Abstract
The remodeling of calcium homeostasis contributes to the cancer hallmarks and the molecular mechanisms involved in calcium channel regulation in tumors remain to be characterized. Here, we report that SigmaR1, a stress-activated chaperone, is required to increase calcium influx by triggering the coupling between SK3, a Ca2+-activated K+ channel (KCNN3) and the voltage-independent calcium channel Orai1. We show that SigmaR1 physically binds SK3 in BC cells. Inhibition of SigmaR1 activity, either by molecular silencing or by the use of sigma ligand (igmesine), decreased SK3 current and Ca2+ entry in breast cancer (BC) and colorectal cancer (CRC) cells. Interestingly, SigmaR1 inhibition diminished SK3 and/or Orai1 levels in lipid nanodomains isolated from BC cells. Analyses of tissue microarray from CRC patients showed higher SigmaR1 expression levels in cancer samples and a correlation with tumor grade. Moreover, the exploration of a cohort of 4937 BC patients indicated that high expression of SigmaR1 and Orai1 channels was significantly correlated to a lower overall survival. As the SK3/Orai1 tandem drives invasive process in CRC and bone metastasis progression in BC, our results may inaugurate innovative therapeutic approaches targeting SigmaR1 to control the remodeling of Ca2+ homeostasis in epithelial cancers.
Collapse
Affiliation(s)
- M Gueguinou
- Inserm-University U1069 Nutrition, Croissance et Cancer, Tours, France
| | - D Crottès
- Université Côte d'Azur, CNRS, Inserm, iBV, France
| | - A Chantôme
- Inserm-University U1069 Nutrition, Croissance et Cancer, Tours, France
| | | | | | - L Clarysse
- Inserm-University U1069 Nutrition, Croissance et Cancer, Tours, France
| | - A Girault
- Inserm-University U1069 Nutrition, Croissance et Cancer, Tours, France
| | - Y Fourbon
- Inserm-University U1069 Nutrition, Croissance et Cancer, Tours, France
| | - P Jézéquel
- Unité de Bioinfomique, Institut de Cancérologie de L'Ouest - René Gauducheau, Centre de Recherche en Cancérologie, UMR-INSERM 892, St Herblain, France
| | - C Guérin-Charbonnel
- Unité de Bioinfomique, Institut de Cancérologie de L'Ouest - René Gauducheau, Centre de Recherche en Cancérologie, UMR-INSERM 892, St Herblain, France
| | - G Fromont
- Inserm-University U1069 Nutrition, Croissance et Cancer, Tours, France
- Service d'Anatomie Pathologique, Hopital Bretonneau, CHRU Tours, Tours, France
| | - P Martin
- Université Côte d'Azur, CNRS, Inserm, iBV, France
| | - B Pellissier
- Université Côte d'Azur, CNRS, Inserm, iBV, France
| | - R Schiappa
- Unité d'Epidémiologie et Biostatistiques (UEB), Centre Antoine Lacassagne, Nice, France
| | - E Chamorey
- Unité d'Epidémiologie et Biostatistiques (UEB), Centre Antoine Lacassagne, Nice, France
| | - O Mignen
- Department of Pathology, Inserm U1078, Brest University Hospital, Brest, France
| | - A Uguen
- Department of Pathology, Inserm U1078, Brest University Hospital, Brest, France
| | - F Borgese
- Université Côte d'Azur, CNRS, Inserm, iBV, France
| | - C Vandier
- Inserm-University U1069 Nutrition, Croissance et Cancer, Tours, France
| | - O Soriani
- Université Côte d'Azur, CNRS, Inserm, iBV, France
| |
Collapse
|
82
|
Webster CP, Smith EF, Shaw PJ, De Vos KJ. Protein Homeostasis in Amyotrophic Lateral Sclerosis: Therapeutic Opportunities? Front Mol Neurosci 2017; 10:123. [PMID: 28512398 PMCID: PMC5411428 DOI: 10.3389/fnmol.2017.00123] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2017] [Accepted: 04/11/2017] [Indexed: 12/11/2022] Open
Abstract
Protein homeostasis (proteostasis), the correct balance between production and degradation of proteins, is essential for the health and survival of cells. Proteostasis requires an intricate network of protein quality control pathways (the proteostasis network) that work to prevent protein aggregation and maintain proteome health throughout the lifespan of the cell. Collapse of proteostasis has been implicated in the etiology of a number of neurodegenerative diseases, including amyotrophic lateral sclerosis (ALS), the most common adult onset motor neuron disorder. Here, we review the evidence linking dysfunctional proteostasis to the etiology of ALS and discuss how ALS-associated insults affect the proteostasis network. Finally, we discuss the potential therapeutic benefit of proteostasis network modulation in ALS.
Collapse
Affiliation(s)
- Christopher P Webster
- Sheffield Institute for Translational Neuroscience (SITraN), Department of Neuroscience, University of SheffieldSheffield, UK
| | - Emma F Smith
- Sheffield Institute for Translational Neuroscience (SITraN), Department of Neuroscience, University of SheffieldSheffield, UK
| | - Pamela J Shaw
- Sheffield Institute for Translational Neuroscience (SITraN), Department of Neuroscience, University of SheffieldSheffield, UK
| | - Kurt J De Vos
- Sheffield Institute for Translational Neuroscience (SITraN), Department of Neuroscience, University of SheffieldSheffield, UK
| |
Collapse
|
83
|
Jesse CM, Bushuven E, Tripathi P, Chandrasekar A, Simon CM, Drepper C, Yamoah A, Dreser A, Katona I, Johann S, Beyer C, Wagner S, Grond M, Nikolin S, Anink J, Troost D, Sendtner M, Goswami A, Weis J. ALS-Associated Endoplasmic Reticulum Proteins in Denervated Skeletal Muscle: Implications for Motor Neuron Disease Pathology. Brain Pathol 2017; 27:781-794. [PMID: 27790792 DOI: 10.1111/bpa.12453] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2016] [Accepted: 10/25/2016] [Indexed: 12/14/2022] Open
Abstract
Alpha-motoneurons and muscle fibres are structurally and functionally interdependent. Both cell types particularly rely on endoplasmic reticulum (ER/SR) functions. Mutations of the ER proteins VAPB, SigR1 and HSP27 lead to hereditary motor neuron diseases (MNDs). Here, we determined the expression profile and localization of these ER proteins/chaperons by immunohistochemistry and immunoblotting in biopsy and autopsy muscle tissue of patients with amyotrophic lateral sclerosis (ALS) and other neurogenic muscular atrophies (NMAs) and compared these patterns to mouse models of neurogenic muscular atrophy. Postsynaptic neuromuscular junction staining for VAPB was intense in normal human and mouse muscle and decreased in denervated Nmd2J mouse muscle fibres. In contrast, VAPB levels together with other chaperones and autophagy markers were increased in extrasynaptic regions of denervated muscle fibres of patients with MNDs and other NMAs, especially at sites of focal myofibrillar disintegration (targets). These findings did not differ between NMAs due to ALS and other causes. G93A-SOD1 mouse muscle fibres showed a similar pattern of protein level increases in denervated muscle fibres. In addition, they showed globular VAPB-immunoreactive structures together with misfolded SOD1 protein accumulations, suggesting a primary myopathic change. Our findings indicate that altered expression and localization of these ER proteins and autophagy markers are part of the dynamic response of muscle fibres to denervation. The ER is particularly prominent and vulnerable in both muscle fibres and alpha-motoneurons. Thus, ER pathology could contribute to the selective build-up of degenerative changes in the neuromuscular axis in MNDs.
Collapse
Affiliation(s)
- C M Jesse
- Institute of Neuropathology, RWTH Aachen University Medical School, Pauwelsstr. 30, 52074 Aachen, Germany.,Department of Neurosurgery, RWTH Aachen University Medical School, Pauwelsstr. 30, 52074 Aachen, Germany
| | - E Bushuven
- Institute of Neuropathology, RWTH Aachen University Medical School, Pauwelsstr. 30, 52074 Aachen, Germany
| | - P Tripathi
- Institute of Neuropathology, RWTH Aachen University Medical School, Pauwelsstr. 30, 52074 Aachen, Germany
| | - A Chandrasekar
- Institute of Neuropathology, RWTH Aachen University Medical School, Pauwelsstr. 30, 52074 Aachen, Germany.,Department of Neurology, Ulm University, Helmholtzstr 8/2, Ulm, 89081, Germany
| | - C M Simon
- Institute of Clinical Neurobiology, University of Würzburg, Versbacherstr. 5, Würzburg, 97078, Germany.,Columbia University Medical Center, Center for Motor Neuron Biology and Disease, 630 West 168th Street, New York, NY, 10032
| | - C Drepper
- Institute of Clinical Neurobiology, University of Würzburg, Versbacherstr. 5, Würzburg, 97078, Germany.,Department of Child and Adolescent Psychiatry, University Hospital Würzburg, Füchsleinstr. 15, Würzburg, 97080, Germany
| | - A Yamoah
- Institute of Neuropathology, RWTH Aachen University Medical School, Pauwelsstr. 30, 52074 Aachen, Germany
| | - A Dreser
- Institute of Neuropathology, RWTH Aachen University Medical School, Pauwelsstr. 30, 52074 Aachen, Germany
| | - I Katona
- Institute of Neuropathology, RWTH Aachen University Medical School, Pauwelsstr. 30, 52074 Aachen, Germany
| | - S Johann
- Institute of Neuroanatomy, RWTH Aachen University Medical School, Pauwelsstr. 30, 52074 Aachen, Germany
| | - C Beyer
- Institute of Neuroanatomy, RWTH Aachen University Medical School, Pauwelsstr. 30, 52074 Aachen, Germany
| | - S Wagner
- Department of Neurology, District Hospital Siegen, Siegen, 57076, Germany
| | - M Grond
- Department of Neurology, District Hospital Siegen, Siegen, 57076, Germany
| | - S Nikolin
- Institute of Neuropathology, RWTH Aachen University Medical School, Pauwelsstr. 30, 52074 Aachen, Germany
| | - J Anink
- Academic Medical Centre, Meibergdreef 9, 1105, AZ, Amsterdam, The Netherlands
| | - D Troost
- Academic Medical Centre, Meibergdreef 9, 1105, AZ, Amsterdam, The Netherlands
| | - M Sendtner
- Institute of Clinical Neurobiology, University of Würzburg, Versbacherstr. 5, Würzburg, 97078, Germany
| | - A Goswami
- Institute of Neuropathology, RWTH Aachen University Medical School, Pauwelsstr. 30, 52074 Aachen, Germany
| | - J Weis
- Institute of Neuropathology, RWTH Aachen University Medical School, Pauwelsstr. 30, 52074 Aachen, Germany
| |
Collapse
|
84
|
Ezawa T, Kawashima Y, Noguchi T, Jung S, Imai N. Convenient synthesis of memantine analogues containing a chiral cyclopropane skeleton as a sigma-1 receptor agonist. ACTA ACUST UNITED AC 2017. [DOI: 10.1016/j.tetasy.2016.12.010] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
85
|
De Mario A, Quintana-Cabrera R, Martinvalet D, Giacomello M. (Neuro)degenerated Mitochondria-ER contacts. Biochem Biophys Res Commun 2017; 483:1096-1109. [DOI: 10.1016/j.bbrc.2016.07.056] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Accepted: 07/10/2016] [Indexed: 01/24/2023]
|
86
|
Casanovas A, Salvany S, Lahoz V, Tarabal O, Piedrafita L, Sabater R, Hernández S, Calderó J, Esquerda JE. Neuregulin 1-ErbB module in C-bouton synapses on somatic motor neurons: molecular compartmentation and response to peripheral nerve injury. Sci Rep 2017; 7:40155. [PMID: 28065942 PMCID: PMC5220293 DOI: 10.1038/srep40155] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2016] [Accepted: 12/02/2016] [Indexed: 12/30/2022] Open
Abstract
The electric activity of lower motor neurons (MNs) appears to play a role in determining cell-vulnerability in MN diseases. MN excitability is modulated by cholinergic inputs through C-type synaptic boutons, which display an endoplasmic reticulum-related subsurface cistern (SSC) adjacent to the postsynaptic membrane. Besides cholinergic molecules, a constellation of proteins involved in different signal-transduction pathways are clustered at C-type synaptic sites (M2 muscarinic receptors, Kv2.1 potassium channels, Ca2+ activated K+ [SK] channels, and sigma-1 receptors [S1R]), but their collective functional significance so far remains unknown. We have previously suggested that neuregulin-1 (NRG1)/ErbBs-based retrograde signalling occurs at this synapse. To better understand signalling through C-boutons, we performed an analysis of the distribution of C-bouton-associated signalling proteins. We show that within SSC, S1R, Kv2.1 and NRG1 are clustered in highly specific, non-overlapping, microdomains, whereas ErbB2 and ErbB4 are present in the adjacent presynaptic compartment. This organization may define highly ordered and spatially restricted sites for different signal-transduction pathways. SSC associated proteins are disrupted in axotomised MNs together with the activation of microglia, which display a positive chemotactism to C-bouton sites. This indicates that C-bouton associated molecules are also involved in neuroinflammatory signalling in diseased MNs, emerging as new potential therapeutic targets.
Collapse
Affiliation(s)
- Anna Casanovas
- Departament de Medicina Experimental, Patologia Neuromuscular Experimental, Facultat de Medicina, Universitat de Lleida/IRBLLEIDA, Av. Rovira Roure 80, 25198 Lleida, Catalonia, Spain
| | - Sara Salvany
- Departament de Medicina Experimental, Patologia Neuromuscular Experimental, Facultat de Medicina, Universitat de Lleida/IRBLLEIDA, Av. Rovira Roure 80, 25198 Lleida, Catalonia, Spain
| | - Víctor Lahoz
- Departament de Medicina Experimental, Patologia Neuromuscular Experimental, Facultat de Medicina, Universitat de Lleida/IRBLLEIDA, Av. Rovira Roure 80, 25198 Lleida, Catalonia, Spain
| | - Olga Tarabal
- Departament de Medicina Experimental, Patologia Neuromuscular Experimental, Facultat de Medicina, Universitat de Lleida/IRBLLEIDA, Av. Rovira Roure 80, 25198 Lleida, Catalonia, Spain
| | - Lídia Piedrafita
- Departament de Medicina Experimental, Patologia Neuromuscular Experimental, Facultat de Medicina, Universitat de Lleida/IRBLLEIDA, Av. Rovira Roure 80, 25198 Lleida, Catalonia, Spain
| | - Raimundo Sabater
- Departament de Medicina Experimental, Patologia Neuromuscular Experimental, Facultat de Medicina, Universitat de Lleida/IRBLLEIDA, Av. Rovira Roure 80, 25198 Lleida, Catalonia, Spain
| | - Sara Hernández
- Departament de Medicina Experimental, Patologia Neuromuscular Experimental, Facultat de Medicina, Universitat de Lleida/IRBLLEIDA, Av. Rovira Roure 80, 25198 Lleida, Catalonia, Spain
| | - Jordi Calderó
- Departament de Medicina Experimental, Patologia Neuromuscular Experimental, Facultat de Medicina, Universitat de Lleida/IRBLLEIDA, Av. Rovira Roure 80, 25198 Lleida, Catalonia, Spain
| | - Josep E. Esquerda
- Departament de Medicina Experimental, Patologia Neuromuscular Experimental, Facultat de Medicina, Universitat de Lleida/IRBLLEIDA, Av. Rovira Roure 80, 25198 Lleida, Catalonia, Spain
| |
Collapse
|
87
|
Nguyen L, Lucke-Wold BP, Mookerjee S, Kaushal N, Matsumoto RR. Sigma-1 Receptors and Neurodegenerative Diseases: Towards a Hypothesis of Sigma-1 Receptors as Amplifiers of Neurodegeneration and Neuroprotection. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 964:133-152. [PMID: 28315269 PMCID: PMC5500918 DOI: 10.1007/978-3-319-50174-1_10] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Sigma-1 receptors are molecular chaperones that may act as pathological mediators and targets for novel therapeutic applications in neurodegenerative diseases. Accumulating evidence indicates that sigma-1 ligands can either directly or indirectly modulate multiple neurodegenerative processes, including excitotoxicity, calcium dysregulation, mitochondrial and endoplasmic reticulum dysfunction, inflammation, and astrogliosis. In addition, sigma-1 ligands may act as disease-modifying agents in the treatment for central nervous system (CNS) diseases by promoting the activity of neurotrophic factors and neural plasticity. Here, we summarize their neuroprotective and neurorestorative effects in different animal models of acute brain injury and chronic neurodegenerative diseases, and highlight their potential role in mitigating disease. Notably, current data suggest that sigma-1 receptor dysfunction worsens disease progression, whereas enhancement amplifies pre-existing functional mechanisms of neuroprotection and/or restoration to slow disease progression. Collectively, the data support a model of the sigma-1 receptor as an amplifier of intracellular signaling, and suggest future clinical applications of sigma-1 ligands as part of multi-therapy approaches to treat neurodegenerative diseases.
Collapse
Affiliation(s)
- Linda Nguyen
- Department of Behavioral Medicine and Psychiatry, School of Medicine, West Virginia University, 930 Chestnut Ridge Road, Morgantown, WV, 26506, USA
- Department of Pharmaceutical Sciences, School of Pharmacy, One Medical Center, West Virginia University, Morgantown, WV, 26506, USA
| | - Brandon P Lucke-Wold
- Graduate Program in Neuroscience, School of Medicine, West Virginia University, One Medical Center Drive, Morgantown, WV, 26506, USA
| | - Shona Mookerjee
- College of Pharmacy, Touro University California, 1310 Club Drive, Vallejo, CA, 94592, USA
| | | | - Rae R Matsumoto
- Department of Behavioral Medicine and Psychiatry, School of Medicine, West Virginia University, 930 Chestnut Ridge Road, Morgantown, WV, 26506, USA.
- College of Pharmacy, Touro University California, 1310 Club Drive, Vallejo, CA, 94592, USA.
| |
Collapse
|
88
|
Mancuso R, Navarro X. Sigma-1 Receptor in Motoneuron Disease. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 964:235-254. [PMID: 28315275 DOI: 10.1007/978-3-319-50174-1_16] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Amyotrophic Lateral Sclerosis (ALS ) is a neurodegenerative disease affecting spinal cord and brain motoneurons , leading to paralysis and early death. Multiple etiopathogenic mechanisms appear to contribute in the development of ALS , including glutamate excitotoxicity, oxidative stress , protein misfolding, mitochondrial defects, impaired axonal transport, inflammation and glial cell alterations. The Sigma-1 receptor is highly expressed in motoneurons of the spinal cord, particularly enriched in the endoplasmic reticulum (ER) at postsynaptic cisternae of cholinergic C-terminals. Several evidences point to participation of Sigma-1R alterations in motoneuron degeneration. Thus, mutations of the transmembrane domain of the Sigma-1R have been described in familial ALS cases. Interestingly, Sigma-1R KO mice display muscle weakness and motoneuron loss. On the other hand, Sigma-1R agonists promote neuroprotection and neurite elongation through activation of protein kinase C on motoneurons in vitro and in vivo after ventral root avulsion. Remarkably, treatment of SOD1 mice, the most usual animal model of ALS , with Sigma-1R agonists resulted in significantly enhanced motoneuron function and preservation, and increased animal survival. Sigma-1R activation also reduced microglial reactivity and increased the glial expression of neurotrophic factors. Two main interconnected mechanisms seem to underlie the effects of Sigma-1R manipulation on motoneurons: modulation of neuronal excitability and regulation of calcium homeostasis. In addition, Sigma-1R also contributes to regulating protein degradation, and reducing oxidative stress. Therefore, the multi-functional nature of the Sigma-1R represents an attractive target for treating aspects of ALS and other motoneuron diseases .
Collapse
Affiliation(s)
- Renzo Mancuso
- Center for Biological Sciences, University of Southampton, Southampton General Hospital, SO16 6YD, Southampton, UK
| | - Xavier Navarro
- Institute of Neurosciences and Department of Cell Biology, Physiology and Immunology, Universitat Autònoma de Barcelona, and Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Bellaterra, Spain.
| |
Collapse
|
89
|
Soriani O, Rapetti-Mauss R. Sigma 1 Receptor and Ion Channel Dynamics in Cancer. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 964:63-77. [PMID: 28315265 DOI: 10.1007/978-3-319-50174-1_6] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
SigmaR1 is a multitasking chaperone protein which has mainly been studied in CNS physiological and pathophysiological processes such as pain, memory, neurodegenerative diseases (amyotrophic lateral sclerosis , Parkinson's and Alzheimer's diseases, retinal neurodegeneration ), stroke and addiction . Strikingly, G-protein and ion channels are the main client protein fami lies of this atypical chaperone and the recent advances that have been performed for the last 10 years demonstrate that SigmaR1 is principally activated following tissue injury and disease development to promote cell survival. In this chapter, we synthesize the data enhancing our comprehension of the interaction between SigmaR1 and ion channels and the unexpected consequences of such functional coupling in cancer development. We also describe a model in which the pro-survival functions of SigmaR1 observed in CNS pathologies are hijacked by cancer cells to shape their electrical signature and behavior in response to the tumor microenvironment .
Collapse
Affiliation(s)
- Olivier Soriani
- University of Nice Sophia Antipolis, CNRS, Inserm, iBV, 06108, Nice, France.
- Bâtiment Sciences Naturelles; UFR Sciences, 06108, Nice, France.
| | | |
Collapse
|
90
|
Cai Y, Yang L, Niu F, Liao K, Buch S. Role of Sigma-1 Receptor in Cocaine Abuse and Neurodegenerative Disease. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 964:163-175. [PMID: 28315271 DOI: 10.1007/978-3-319-50174-1_12] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Sigma-1 receptors (Sig-1R) are recognized as a unique class of non-G protein-coupled intracellular protein. Sig-1R binds to its ligand such as cocaine , resulting in dissociation of Sig-1R from mitochondrion-associated ER membrane (MAM) to the endoplasmic reticulum (ER), plasma membrane, and nuclear membrane, regulating function of various proteins. Sig-1R has diverse roles in both physiological as well as in pathogenic processes. The disruption of Sig-1R pathways has been implicated as causative mechanism(s) in the development of both neurodegenerative disorders such as Alzheimer disease (AD ), Parkinson disease (PD ), amyotrophic lateral sclerosis (ALS ) and Huntington Disease (HD ) . Additionally, the interaction of cocaine and Sig-1R has more recently been implicated in potentiating the pathogenesis of HIV-associated neurocognitive disorders (HAND) through impairment of blood-brain barrier (BBB), microglial activation and astrogliosis. On the other hand, restoration of Sig-1R homeostasis has been shown to exert neuroprotective effects. In this review, we provide an overview of how Sig-1R plays a role in the pathogenesis of neurodegenerative disorders and cocaine and implications for future development of therapeutic strategies.
Collapse
Affiliation(s)
- Yu Cai
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, 985880 Nebraska Medical Center, DRC 8024, Omaha, NE, 68198-5880, USA
| | - Lu Yang
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, 985880 Nebraska Medical Center, DRC 8024, Omaha, NE, 68198-5880, USA
| | - Fang Niu
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, 985880 Nebraska Medical Center, DRC 8024, Omaha, NE, 68198-5880, USA
| | - Ke Liao
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, 985880 Nebraska Medical Center, DRC 8024, Omaha, NE, 68198-5880, USA
| | - Shilpa Buch
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, 985880 Nebraska Medical Center, DRC 8024, Omaha, NE, 68198-5880, USA.
| |
Collapse
|
91
|
Hattori N, Arano T, Hatano T, Mori A, Imai Y. Mitochondrial-Associated Membranes in Parkinson's Disease. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 997:157-169. [PMID: 28815529 DOI: 10.1007/978-981-10-4567-7_12] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Parkinson's disease (PD) is a common neurodegenerative disorder, with ageing being a major risk factor. Accordingly, estimates predict an increasing number of PD patients due to our expanding life span. Consequently, developing a true disease-modifying therapy is necessary. In this regard, monogenic PD offers a suitable means for determining pathogenesis. Among monogenic forms of PD, mitochondrial dysfunction may be a major cause and is also likely to be involved in sporadic PD. Thus, mitochondrial impairment may be a common pathway. Recently, mitochondria-associated membranes (MAM) were identified as dynamic sites between mitochondria and endoplasmic reticulum. Indeed, the gene product of α-synuclein is a major component of MAM, with other gene products also involved. This review focuses on the possibility of using MAM as novel therapeutic targets.
Collapse
Affiliation(s)
- Nobutaka Hattori
- Department of Neurology, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo, Tokyo, 113-8421, Japan.
| | - Taku Arano
- Department of Neurology, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo, Tokyo, 113-8421, Japan
| | - Taku Hatano
- Department of Neurology, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo, Tokyo, 113-8421, Japan
| | - Akio Mori
- Department of Neurology, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo, Tokyo, 113-8421, Japan
| | - Yuzuru Imai
- Department of Neurology, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo, Tokyo, 113-8421, Japan
| |
Collapse
|
92
|
Watanabe S, Ilieva H, Tamada H, Nomura H, Komine O, Endo F, Jin S, Mancias P, Kiyama H, Yamanaka K. Mitochondria-associated membrane collapse is a common pathomechanism in SIGMAR1- and SOD1-linked ALS. EMBO Mol Med 2016; 8:1421-1437. [PMID: 27821430 PMCID: PMC5167132 DOI: 10.15252/emmm.201606403] [Citation(s) in RCA: 189] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2016] [Revised: 09/21/2016] [Accepted: 09/27/2016] [Indexed: 01/11/2023] Open
Abstract
A homozygous mutation in the gene for sigma 1 receptor (Sig1R) is a cause of inherited juvenile amyotrophic lateral sclerosis (ALS16). Sig1R localizes to the mitochondria-associated membrane (MAM), which is an interface of mitochondria and endoplasmic reticulum. However, the role of the MAM in ALS is not fully elucidated. Here, we identified a homozygous p.L95fs mutation of Sig1R as a novel cause of ALS16. ALS-linked Sig1R variants were unstable and incapable of binding to inositol 1,4,5-triphosphate receptor type 3 (IP3R3). The onset of mutant Cu/Zn superoxide dismutase (SOD1)-mediated ALS disease in mice was accelerated when Sig1R was deficient. Moreover, either deficiency of Sig1R or accumulation of mutant SOD1 induced MAM disruption, resulting in mislocalization of IP3R3 from the MAM, calpain activation, and mitochondrial dysfunction. Our findings indicate that a loss of Sig1R function is causative for ALS16, and collapse of the MAM is a common pathomechanism in both Sig1R- and SOD1-linked ALS Furthermore, our discovery of the selective enrichment of IP3R3 in motor neurons suggests that integrity of the MAM is crucial for the selective vulnerability in ALS.
Collapse
Affiliation(s)
- Seiji Watanabe
- Department of Neuroscience and Pathobiology, Research Institute of Environmental Medicine, Nagoya University, Nagoya, Aichi, Japan
| | - Hristelina Ilieva
- Houston Methodist Hospital, Houston, TX, USA
- Department of Neurology, Johns Hopkins University, Baltimore, MD, USA
| | - Hiromi Tamada
- Department of Functional Anatomy and Neuroscience, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
| | - Hanae Nomura
- Department of Neuroscience and Pathobiology, Research Institute of Environmental Medicine, Nagoya University, Nagoya, Aichi, Japan
| | - Okiru Komine
- Department of Neuroscience and Pathobiology, Research Institute of Environmental Medicine, Nagoya University, Nagoya, Aichi, Japan
| | - Fumito Endo
- Department of Neuroscience and Pathobiology, Research Institute of Environmental Medicine, Nagoya University, Nagoya, Aichi, Japan
| | - Shijie Jin
- Department of Neuroscience and Pathobiology, Research Institute of Environmental Medicine, Nagoya University, Nagoya, Aichi, Japan
| | - Pedro Mancias
- Department of Pediatrics, The University of Texas Medical School at Houston, Houston, TX, USA
| | - Hiroshi Kiyama
- Department of Functional Anatomy and Neuroscience, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
| | - Koji Yamanaka
- Department of Neuroscience and Pathobiology, Research Institute of Environmental Medicine, Nagoya University, Nagoya, Aichi, Japan
| |
Collapse
|
93
|
Brauers E, Roos A, Kollipara L, Zahedi RP, Beckmann A, Mohanadas N, Bauer H, Häusler M, Thoma S, Kress W, Senderek J, Weis J. The Caveolin-3 G56S sequence variant of unknown significance: Muscle biopsy findings and functional cell biological analysis. Proteomics Clin Appl 2016; 11. [PMID: 27739254 PMCID: PMC5248598 DOI: 10.1002/prca.201600007] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2016] [Revised: 08/09/2016] [Accepted: 10/11/2016] [Indexed: 12/31/2022]
Abstract
Purpose In the era of next‐generation sequencing, we are increasingly confronted with sequence variants of unknown significance. This phenomenon is also known for variations in Caveolin‐3 and can complicate the molecular diagnosis of the disease. Here, we aimed to study the ambiguous character of the G56S Caveolin‐3 variant. Experimental design A comprehensive approach combining genetic and morphological studies of muscle derived from carriers of the G56S Caveolin‐3 variant were carried out and linked to biochemical assays (including phosphoblot studies and proteome profiling) and morphological investigations of cultured myoblasts. Results Muscles showed moderate chronic myopathic changes in all carriers of the variant. Myogenic RCMH cells expressing the G56S Caveolin‐3 protein presented irregular Caveolin‐3 deposits within the Golgi in addition to a regular localization of the protein to the plasma membrane. This result was associated with abnormal findings on the ultra‐structural level. Phosphoblot studies revealed that G56S affects EGFR‐signaling. Proteomic profiling demonstrated alterations in levels of physiologically relevant proteins which are indicative for antagonization of G56S Caveolin‐3 expression. Remarkably, some proteomic alterations were enhanced by osmotic/mechanical stress. Conclusions and clinical relevance Our studies suggest that G56S might influence the manifestation of myopathic changes upon the presence of additional cellular stress burden. Results of our studies moreover improve the current understanding of (genetic) causes of myopathic disorders classified as caveolinopathies.
Collapse
Affiliation(s)
- Eva Brauers
- Institute of Neuropathology, RWTH Aachen University Hospital, Aachen, Germany
| | - Andreas Roos
- Institute of Neuropathology, RWTH Aachen University Hospital, Aachen, Germany.,Leibniz-Institut für Analytische Wissenschaften - ISAS e.V, Dortmund, Germany
| | | | - René P Zahedi
- Leibniz-Institut für Analytische Wissenschaften - ISAS e.V, Dortmund, Germany
| | - Alf Beckmann
- Medizinisches Versorgungszentrum Dr. Eberhard und Partner, Dortmund, Germany
| | - Nilane Mohanadas
- Institute of Neuropathology, RWTH Aachen University Hospital, Aachen, Germany
| | - Hartmut Bauer
- Department of Neurology, Marien-Hospital, Euskirchen, Germany
| | - Martin Häusler
- Department of Pediatrics, RWTH Aachen University Hospital, Aachen, Germany
| | | | - Wolfram Kress
- Institute of Human Genetics, University of Würzburg, Würzburg, Germany
| | - Jan Senderek
- Friedrich-Baur-Institut, Neurologische Klinik und Poliklinik, Ludwig-Maximilians-University, München, Germany
| | - Joachim Weis
- Institute of Neuropathology, RWTH Aachen University Hospital, Aachen, Germany
| |
Collapse
|
94
|
Recent Advances in Neurogenic Small Molecules as Innovative Treatments for Neurodegenerative Diseases. Molecules 2016; 21:molecules21091165. [PMID: 27598108 PMCID: PMC6273783 DOI: 10.3390/molecules21091165] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2016] [Revised: 08/22/2016] [Accepted: 08/29/2016] [Indexed: 12/20/2022] Open
Abstract
The central nervous system of adult mammals has long been considered as a complex static structure unable to undergo any regenerative process to refurbish its dead nodes. This dogma was challenged by Altman in the 1960s and neuron self-renewal has been demonstrated ever since in many species, including humans. Aging, neurodegenerative, and some mental diseases are associated with an exponential decrease in brain neurogenesis. Therefore, the controlled pharmacological stimulation of the endogenous neural stem cells (NSCs) niches might counteract the neuronal loss in Alzheimer’s disease (AD) and other pathologies, opening an exciting new therapeutic avenue. In the last years, druggable molecular targets and signalling pathways involved in neurogenic processes have been identified, and as a consequence, different drug types have been developed and tested in neuronal plasticity. This review focuses on recent advances in neurogenic agents acting at serotonin and/or melatonin systems, Wnt/β-catenin pathway, sigma receptors, nicotinamide phosphoribosyltransferase (NAMPT) and nuclear erythroid 2-related factor (Nrf2).
Collapse
|
95
|
Gregianin E, Pallafacchina G, Zanin S, Crippa V, Rusmini P, Poletti A, Fang M, Li Z, Diano L, Petrucci A, Lispi L, Cavallaro T, Fabrizi GM, Muglia M, Boaretto F, Vettori A, Rizzuto R, Mostacciuolo ML, Vazza G. Loss-of-function mutations in the SIGMAR1 gene cause distal hereditary motor neuropathy by impairing ER-mitochondria tethering and Ca2+ signalling. Hum Mol Genet 2016; 25:3741-3753. [PMID: 27402882 DOI: 10.1093/hmg/ddw220] [Citation(s) in RCA: 86] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2016] [Revised: 06/29/2016] [Accepted: 06/30/2016] [Indexed: 01/14/2023] Open
Abstract
Distal hereditary motor neuropathies (dHMNs) are clinically and genetically heterogeneous neurological conditions characterized by degeneration of the lower motor neurons. So far, 18 dHMN genes have been identified, however, about 80% of dHMN cases remain without a molecular diagnosis. By a combination of autozygosity mapping, identity-by-descent segment detection and whole-exome sequencing approaches, we identified two novel homozygous mutations in the SIGMAR1 gene (p.E138Q and p.E150K) in two distinct Italian families affected by an autosomal recessive form of HMN. Functional analyses in several neuronal cell lines strongly support the pathogenicity of the mutations and provide insights into the underlying pathomechanisms involving the regulation of ER-mitochondria tethering, Ca2+ homeostasis and autophagy. Indeed, in vitro, both mutations reduce cell viability, the formation of abnormal protein aggregates preventing the correct targeting of sigma-1R protein to the mitochondria-associated ER membrane (MAM) and thus impinging on the global Ca2+ signalling. Our data definitively demonstrate the involvement of SIGMAR1 in motor neuron maintenance and survival by correlating, for the first time in the Caucasian population, mutations in this gene to distal motor dysfunction and highlight the chaperone activity of sigma-1R at the MAM as a critical aspect in dHMN pathology.
Collapse
Affiliation(s)
| | - Giorgia Pallafacchina
- Department of Biomedical Sciences, University of Padova and CNR Neuroscience Institute, Padova, Italy
| | - Sofia Zanin
- Department of Biomedical Sciences, University of Padova and CNR Neuroscience Institute, Padova, Italy
| | - Valeria Crippa
- Experimental Neurobiology Lab, IRCCS "C. Mondino" National Neurological Institute, Pavia, Italy
| | - Paola Rusmini
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy
| | - Angelo Poletti
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy
| | - Mingyan Fang
- Department of Science & Technology, BGI-Shenzhen, Shenzhen, China
| | - Zhouxuan Li
- Department of Science & Technology, BGI-Shenzhen, Shenzhen, China
| | - Laura Diano
- Medical Genetics, University Hospital "Tor Vergata", Roma, Italy
| | - Antonio Petrucci
- Neuromuscular and Rare Neurological Diseases Centre Neurology & Neurophysiopathology Unit, ASO San Camillo-Forlanini Hospital of Rome, Rome, Italy
| | - Ludovico Lispi
- Neuromuscular and Rare Neurological Diseases Centre Neurology & Neurophysiopathology Unit, ASO San Camillo-Forlanini Hospital of Rome, Rome, Italy
| | - Tiziana Cavallaro
- Section of Neuropathology, Neurological and Movement Sciences, University of Verona, Verona, Italy
| | - Gian M Fabrizi
- Section of Neuropathology, Neurological and Movement Sciences, University of Verona, Verona, Italy
| | - Maria Muglia
- CNR Institute of Neurological Sciences, Mangone, Cosenza, Italy
| | | | | | - Rosario Rizzuto
- Department of Biomedical Sciences, University of Padova and CNR Neuroscience Institute, Padova, Italy
| | | | | |
Collapse
|
96
|
Zufiría M, Gil-Bea FJ, Fernández-Torrón R, Poza JJ, Muñoz-Blanco JL, Rojas-García R, Riancho J, López de Munain A. ALS: A bucket of genes, environment, metabolism and unknown ingredients. Prog Neurobiol 2016; 142:104-129. [DOI: 10.1016/j.pneurobio.2016.05.004] [Citation(s) in RCA: 113] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2015] [Revised: 03/22/2016] [Accepted: 05/09/2016] [Indexed: 12/11/2022]
|
97
|
Su TP, Su TC, Nakamura Y, Tsai SY. The Sigma-1 Receptor as a Pluripotent Modulator in Living Systems. Trends Pharmacol Sci 2016; 37:262-278. [PMID: 26869505 PMCID: PMC4811735 DOI: 10.1016/j.tips.2016.01.003] [Citation(s) in RCA: 237] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2015] [Revised: 01/03/2016] [Accepted: 01/05/2016] [Indexed: 01/21/2023]
Abstract
The sigma-1 receptor (Sig-1R) is an endoplasmic reticulum (ER) protein that resides specifically in the mitochondria-associated endoplasmic reticulum (ER) membrane (MAM), an interface between ER and mitochondria. In addition to being able to translocate to the plasma membrane (PM) to interact with ion channels and other receptors, Sig-1R also occurs at the nuclear envelope, where it recruits chromatin-remodeling factors to affect the transcription of genes. Sig-1Rs have also been reported to interact with other membranous or soluble proteins at other loci, including the cytosol, and to be involved in several central nervous system (CNS) diseases. Here, we propose that Sig-1R is a pluripotent modulator with resultant multiple functional manifestations in living systems.
Collapse
Affiliation(s)
- Tsung-Ping Su
- Cellular Pathobiology Section, Integrative Neuroscience Research Branch, Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Department of Health and Human Services, Baltimore, MD 21224, USA.
| | - Tzu-Chieh Su
- Cellular Pathobiology Section, Integrative Neuroscience Research Branch, Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Department of Health and Human Services, Baltimore, MD 21224, USA
| | - Yoki Nakamura
- Cellular Pathobiology Section, Integrative Neuroscience Research Branch, Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Department of Health and Human Services, Baltimore, MD 21224, USA
| | - Shang-Yi Tsai
- Cellular Pathobiology Section, Integrative Neuroscience Research Branch, Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Department of Health and Human Services, Baltimore, MD 21224, USA
| |
Collapse
|
98
|
Krols M, van Isterdael G, Asselbergh B, Kremer A, Lippens S, Timmerman V, Janssens S. Mitochondria-associated membranes as hubs for neurodegeneration. Acta Neuropathol 2016; 131:505-23. [PMID: 26744348 PMCID: PMC4789254 DOI: 10.1007/s00401-015-1528-7] [Citation(s) in RCA: 168] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2015] [Revised: 12/18/2015] [Accepted: 12/18/2015] [Indexed: 12/17/2022]
Abstract
There is a growing appreciation that membrane-bound organelles in eukaryotic cells communicate directly with one another through direct membrane contact sites. Mitochondria-associated membranes are specialized subdomains of the endoplasmic reticulum that function as membrane contact sites between the endoplasmic reticulum and mitochondria. These sites have emerged as major players in lipid metabolism and calcium signaling. More recently also autophagy and mitochondrial dynamics have been found to be regulated at ER-mitochondria contact sites. Neurons critically depend on mitochondria-associated membranes as a means to exchange metabolites and signaling molecules between these organelles. This is underscored by the fact that genes affecting mitochondrial and endoplasmic reticulum homeostasis are clearly overrepresented in several hereditary neurodegenerative disorders. Conversely, the processes affected by the contact sites between the endoplasmic reticulum and mitochondria are widely implicated in neurodegeneration. This review will focus on the most recent data addressing the structural composition and function of the mitochondria-associated membranes. In addition, the 3D morphology of the contact sites as observed using volume electron microscopy is discussed. Finally, it will highlight the role of several key proteins associated with these contact sites that are involved not only in dementias, amyotrophic lateral sclerosis and Parkinson's disease, but also in axonopathies such as hereditary spastic paraplegia and Charcot-Marie-Tooth disease.
Collapse
|
99
|
Kollipara L, Buchkremer S, Weis J, Brauers E, Hoss M, Rütten S, Caviedes P, Zahedi RP, Roos A. Proteome Profiling and Ultrastructural Characterization of the Human RCMH Cell Line: Myoblastic Properties and Suitability for Myopathological Studies. J Proteome Res 2016; 15:945-55. [PMID: 26781476 DOI: 10.1021/acs.jproteome.5b00972] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Studying (neuro)muscular disorders is a major topic in biomedicine with a demand for suitable model systems. Continuous cell culture (in vitro) systems have several technical advantages over in vivo systems and became widely used tools for discovering physiological/pathophysiological mechanisms in muscle. In particular, myoblast cell lines are suitable model systems to study complex biochemical adaptations occurring in skeletal muscle and cellular responses to altered genetic/environmental conditions. Whereas most in vitro studies use extensively characterized murine C2C12 cells, a comprehensive description of an equivalent human cell line, not genetically manipulated for immortalization, is lacking. Therefore, we characterized human immortal myoblastic RCMH cells using scanning (SEM) and transmission electron microscopy (TEM) and proteomics. Among more than 6200 identified proteins we confirm the known expression of proteins important for muscle function. Comparing the RCMH proteome with two well-defined nonskeletal muscle cells lines (HeLa, U2OS) revealed a considerable enrichment of proteins important for muscle function. SEM/TEM confirmed the presence of agglomerates of cytoskeletal components/intermediate filaments and a prominent rough ER. In conclusion, our results indicate RMCH as a suitable in vitro model for investigating muscle function-related processes such as mechanical stress burden and mechanotransduction, EC coupling, cytoskeleton, muscle cell metabolism and development, and (ER-associated) myopathic disorders.
Collapse
Affiliation(s)
- Laxmikanth Kollipara
- Leibniz-Institut für Analytische Wissenschaften, ISAS e.V. , Otto-Hahn-Str. 6b, 44227 Dortmund, Germany
| | - Stephan Buchkremer
- Institute of Neuropathology, RWTH Aachen University Hospital , Pauwelsstrasse 30, D-52074 Aachen, Germany
| | - Joachim Weis
- Institute of Neuropathology, RWTH Aachen University Hospital , Pauwelsstrasse 30, D-52074 Aachen, Germany
| | - Eva Brauers
- Institute of Neuropathology, RWTH Aachen University Hospital , Pauwelsstrasse 30, D-52074 Aachen, Germany
| | - Mareike Hoss
- Electron Microscopic Facility, Institute of Pathology, RWTH Aachen University Hospital , D-52074 Aachen, Germany
| | - Stephan Rütten
- Electron Microscopic Facility, Institute of Pathology, RWTH Aachen University Hospital , D-52074 Aachen, Germany
| | - Pablo Caviedes
- Programa de Farmacología Molecular y Clínica, ICBM, Facultad de Medicina, Universidad de Chile , Santiago 1058, Chile
| | - René P Zahedi
- Leibniz-Institut für Analytische Wissenschaften, ISAS e.V. , Otto-Hahn-Str. 6b, 44227 Dortmund, Germany
| | - Andreas Roos
- Leibniz-Institut für Analytische Wissenschaften, ISAS e.V. , Otto-Hahn-Str. 6b, 44227 Dortmund, Germany.,Institute of Neuropathology, RWTH Aachen University Hospital , Pauwelsstrasse 30, D-52074 Aachen, Germany
| |
Collapse
|
100
|
Crottès D, Rapetti-Mauss R, Alcaraz-Perez F, Tichet M, Gariano G, Martial S, Guizouarn H, Pellissier B, Loubat A, Popa A, Paquet A, Presta M, Tartare-Deckert S, Cayuela ML, Martin P, Borgese F, Soriani O. SIGMAR1 Regulates Membrane Electrical Activity in Response to Extracellular Matrix Stimulation to Drive Cancer Cell Invasiveness. Cancer Res 2016; 76:607-18. [PMID: 26645564 DOI: 10.1158/0008-5472.can-15-1465] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2015] [Accepted: 10/27/2015] [Indexed: 01/12/2023]
Abstract
The sigma 1 receptor (Sig1R) is a stress-activated chaperone that regulates ion channels and is associated with pathologic conditions, such as stroke, neurodegenerative diseases, and addiction. Aberrant expression levels of ion channels and Sig1R have been detected in tumors and cancer cells, such as myeloid leukemia and colorectal cancer, but the link between ion channel regulation and Sig1R overexpression during malignancy has not been established. In this study, we found that Sig1R dynamically controls the membrane expression of the human voltage-dependent K(+) channel human ether-à-go-go-related gene (hERG) in myeloid leukemia and colorectal cancer cell lines. Sig1R promoted the formation of hERG/β1-integrin signaling complexes upon extracellular matrix stimulation, triggering the activation of the PI3K/AKT pathway. Consequently, the presence of Sig1R in cancer cells increased motility and VEGF secretion. In vivo, Sig1R expression enhanced the aggressiveness of tumor cells by potentiating invasion and angiogenesis, leading to poor survival. Collectively, our findings highlight a novel function for Sig1R in mediating cross-talk between cancer cells and their microenvironment, thus driving oncogenesis by shaping cellular electrical activity in response to extracellular signals. Given the involvement of ion channels in promoting several hallmarks of cancer, our study also offers a potential strategy to therapeutically target ion channel function through Sig1R inhibition.
Collapse
Affiliation(s)
- David Crottès
- Université Nice Sophia Antipolis, iBV, Nice, France. CNRS, iBV, UMR7277, Nice, France. INSERM U1091, Nice, France. Department of Physiology, University of California, San Francisco, San Francisco, California
| | - Raphael Rapetti-Mauss
- Université Nice Sophia Antipolis, iBV, Nice, France. CNRS, iBV, UMR7277, Nice, France. INSERM U1091, Nice, France
| | - Francisca Alcaraz-Perez
- Telomerase, Aging and Cancer Group, Research Unit, Department of Surgery, CIBERehd, University Hospital "Virgen de la Arrixaca", Murcia, Spain. Instituto Murciano de Investigación Biosanitaria (IMIB), Murcia, Spain
| | - Mélanie Tichet
- Université Nice Sophia Antipolis, C3M, Inserm U1065, Nice, France
| | - Giuseppina Gariano
- Unit of Oncology and Experimental Immunology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Sonia Martial
- Université Nice Sophia Antipolis, iBV, Nice, France. CNRS, iBV, UMR7277, Nice, France. INSERM U1091, Nice, France
| | - Hélène Guizouarn
- Université Nice Sophia Antipolis, iBV, Nice, France. CNRS, iBV, UMR7277, Nice, France. INSERM U1091, Nice, France
| | - Bernard Pellissier
- Université Nice Sophia Antipolis, iBV, Nice, France. CNRS, iBV, UMR7277, Nice, France. INSERM U1091, Nice, France
| | - Agnès Loubat
- Université Nice Sophia Antipolis, iBV, Nice, France. CNRS, iBV, UMR7277, Nice, France. INSERM U1091, Nice, France
| | - Alexandra Popa
- Université Nice Sophia Antipolis, IPMC, CNRS UMR7275, Sophia Antipolis, France
| | - Agnès Paquet
- Université Nice Sophia Antipolis, IPMC, CNRS UMR7275, Sophia Antipolis, France
| | - Marco Presta
- Unit of Oncology and Experimental Immunology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | | | - Maria Luisa Cayuela
- Telomerase, Aging and Cancer Group, Research Unit, Department of Surgery, CIBERehd, University Hospital "Virgen de la Arrixaca", Murcia, Spain. Instituto Murciano de Investigación Biosanitaria (IMIB), Murcia, Spain
| | - Patrick Martin
- Université Nice Sophia Antipolis, iBV, Nice, France. CNRS, iBV, UMR7277, Nice, France. INSERM U1091, Nice, France
| | - Franck Borgese
- Université Nice Sophia Antipolis, iBV, Nice, France. CNRS, iBV, UMR7277, Nice, France. INSERM U1091, Nice, France
| | - Olivier Soriani
- Université Nice Sophia Antipolis, iBV, Nice, France. CNRS, iBV, UMR7277, Nice, France. INSERM U1091, Nice, France.
| |
Collapse
|