1
|
Amodei L, Ruggieri AG, Potenza F, Viele M, Dufrusine B, Franciotti R, Pietrangelo L, Ardini M, Stuppia L, Federici L, De Laurenzi V, Sallese M. Sil1-deficient fibroblasts generate an aberrant extracellular matrix leading to tendon disorganisation in Marinesco-Sjögren syndrome. J Transl Med 2024; 22:787. [PMID: 39180052 PMCID: PMC11342654 DOI: 10.1186/s12967-024-05582-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Accepted: 08/05/2024] [Indexed: 08/26/2024] Open
Abstract
BACKGROUND Marinesco-Sjögren syndrome (MSS) is an autosomal recessive neuromuscular disorder that arises in early childhood and is characterized by congenital cataracts, myopathy associated with muscle weakness, and degeneration of Purkinje neurons leading to ataxia. About 60% of MSS patients have loss-of-function mutations in the SIL1 gene. Sil1 is an endoplasmic reticulum (ER) protein required for the release of ADP from the master chaperone Bip, which in turn will release the folded proteins. The expression of non-functional Sil1 leads to the accumulation of unfolded proteins in the ER and this triggers the unfolded protein response (UPR). A dysfunctional UPR could be a key element in the pathogenesis of MSS, although our knowledge of the molecular pathology of MSS is still incomplete. METHODS RNA-Seq transcriptomics was analysed using the String database and the Ingenuity Pathway Analysis platform. Fluorescence confocal microscopy was used to study the remodelling of the extracellular matrix (ECM). Transmission electron microscopy (TEM) was used to reveal the morphology of the ECM in vitro and in mouse tendon. RESULTS Our transcriptomic analysis, performed on patient-derived fibroblasts, revealed 664 differentially expressed (DE) transcripts. Enrichment analysis of DE genes confirmed that the patient fibroblasts have a membrane trafficking issue. Furthermore, this analysis indicated that the extracellular space/ECM and the cell adhesion machinery, which together account for around 300 transcripts, could be affected in MSS. Functional assays showed that patient fibroblasts have a reduced capacity of ECM remodelling, reduced motility, and slower spreading during adhesion to Petri dishes. TEM micrographs of negative-stained ECM samples from these fibroblasts show differences of filaments in terms of morphology and size. Finally, structural analysis of the myotendinous junction of the soleus muscle and surrounding regions of the Achilles tendon revealed a disorganization of collagen fibres in the mouse model of MSS (woozy). CONCLUSIONS ECM alterations can affect the proper functioning of several organs, including those damaged in MSS such as the central nervous system, skeletal muscle, bone and lens. On this basis, we propose that aberrant ECM is a key pathological feature of MSS and may help explain most of its clinical manifestations.
Collapse
Affiliation(s)
- Laura Amodei
- Department of Innovative Technologies in Medicine and Dentistry, Chieti, Italy
- Center for Advanced Studies and Technology (CAST), Chieti, Italy
| | - Anna Giulia Ruggieri
- Department of Innovative Technologies in Medicine and Dentistry, Chieti, Italy
- Center for Advanced Studies and Technology (CAST), Chieti, Italy
| | - Francesca Potenza
- Department of Innovative Technologies in Medicine and Dentistry, Chieti, Italy
- Center for Advanced Studies and Technology (CAST), Chieti, Italy
| | - Marianna Viele
- Department of Innovative Technologies in Medicine and Dentistry, Chieti, Italy
- Center for Advanced Studies and Technology (CAST), Chieti, Italy
| | - Beatrice Dufrusine
- Department of Bioscience and Technology for Food Agriculture and Environment, University of Teramo, Teramo, 64100, Italy
| | | | | | - Matteo Ardini
- Department of Life, Health and Environmental Sciences, University of L'Aquila, L'Aquila, 67100, Italy
| | - Liborio Stuppia
- Center for Advanced Studies and Technology (CAST), Chieti, Italy
- Department of Psychological Health and Territorial Sciences, "G. d'Annunzio" University of Chieti-Pescara, Chieti, 66100, Italy
| | - Luca Federici
- Department of Innovative Technologies in Medicine and Dentistry, Chieti, Italy
- Center for Advanced Studies and Technology (CAST), Chieti, Italy
| | - Vincenzo De Laurenzi
- Department of Innovative Technologies in Medicine and Dentistry, Chieti, Italy
- Center for Advanced Studies and Technology (CAST), Chieti, Italy
| | - Michele Sallese
- Department of Innovative Technologies in Medicine and Dentistry, Chieti, Italy.
- Center for Advanced Studies and Technology (CAST), Chieti, Italy.
| |
Collapse
|
2
|
Jennings MJ, Hathazi D, Nguyen CDL, Munro B, Münchberg U, Ahrends R, Schenck A, Eidhof I, Freier E, Synofzik M, Horvath R, Roos A. Intracellular Lipid Accumulation and Mitochondrial Dysfunction Accompanies Endoplasmic Reticulum Stress Caused by Loss of the Co-chaperone DNAJC3. Front Cell Dev Biol 2021; 9:710247. [PMID: 34692675 PMCID: PMC8526738 DOI: 10.3389/fcell.2021.710247] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2021] [Accepted: 09/02/2021] [Indexed: 12/25/2022] Open
Abstract
Recessive mutations in DNAJC3, an endoplasmic reticulum (ER)-resident BiP co-chaperone, have been identified in patients with multisystemic neurodegeneration and diabetes mellitus. To further unravel these pathomechanisms, we employed a non-biased proteomic approach and identified dysregulation of several key cellular pathways, suggesting a pathophysiological interplay of perturbed lipid metabolism, mitochondrial bioenergetics, ER-Golgi function, and amyloid-beta processing. Further functional investigations in fibroblasts of patients with DNAJC3 mutations detected cellular accumulation of lipids and an increased sensitivity to cholesterol stress, which led to activation of the unfolded protein response (UPR), alterations of the ER-Golgi machinery, and a defect of amyloid precursor protein. In line with the results of previous studies, we describe here alterations in mitochondrial morphology and function, as a major contributor to the DNAJC3 pathophysiology. Hence, we propose that the loss of DNAJC3 affects lipid/cholesterol homeostasis, leading to UPR activation, β-amyloid accumulation, and impairment of mitochondrial oxidative phosphorylation.
Collapse
Affiliation(s)
- Matthew J. Jennings
- Department of Clinical Neuroscience, University of Cambridge, Cambridge, United Kingdom
| | - Denisa Hathazi
- Department of Clinical Neuroscience, University of Cambridge, Cambridge, United Kingdom
- Leibniz-Institut für Analytische Wissenschaften – ISAS – e.V., Dortmund, Germany
| | - Chi D. L. Nguyen
- Leibniz-Institut für Analytische Wissenschaften – ISAS – e.V., Dortmund, Germany
| | - Benjamin Munro
- Department of Clinical Neuroscience, University of Cambridge, Cambridge, United Kingdom
| | - Ute Münchberg
- Leibniz-Institut für Analytische Wissenschaften – ISAS – e.V., Dortmund, Germany
| | - Robert Ahrends
- Leibniz-Institut für Analytische Wissenschaften – ISAS – e.V., Dortmund, Germany
| | - Annette Schenck
- Department of Human Genetics, Donders Institute for Brain, Cognition and Behavior, Radboud University Medical Center, Nijmegen, Netherlands
| | - Ilse Eidhof
- Department of Human Genetics, Donders Institute for Brain, Cognition and Behavior, Radboud University Medical Center, Nijmegen, Netherlands
| | - Erik Freier
- Leibniz-Institut für Analytische Wissenschaften – ISAS – e.V., Dortmund, Germany
| | - Matthis Synofzik
- Department of Neurodegenerative Diseases, Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
- German Centre for Neurodegenerative Diseases (DZNE), Tübingen, Germany
| | - Rita Horvath
- Department of Clinical Neuroscience, University of Cambridge, Cambridge, United Kingdom
| | - Andreas Roos
- Leibniz-Institut für Analytische Wissenschaften – ISAS – e.V., Dortmund, Germany
- Department of Pediatric Neurology, Developmental Neurology and Social Pediatrics, Children’s Hospital University of Essen, Essen, Germany
| |
Collapse
|
3
|
Sumya FT, Pokrovskaya ID, Lupashin V. Development and Initial Characterization of Cellular Models for COG Complex-Related CDG-II Diseases. Front Genet 2021; 12:733048. [PMID: 34603392 PMCID: PMC8484713 DOI: 10.3389/fgene.2021.733048] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Accepted: 09/06/2021] [Indexed: 12/12/2022] Open
Abstract
Conserved Oligomeric Golgi (COG) is an octameric protein complex that orchestrates intra-Golgi trafficking of glycosylation enzymes. Over a hundred individuals with 31 different COG mutations have been identified until now. The cellular phenotypes and clinical presentations of COG-CDGs are heterogeneous, and patients primarily represent neurological, skeletal, and hepatic abnormalities. The establishment of a cellular COG disease model will benefit the molecular study of the disease, explaining the detailed sequence of the interplay between the COG complex and the trafficking machinery. Moreover, patient fibroblasts are not a good representative of all the organ systems and cell types that are affected by COG mutations. We developed and characterized cellular models for human COG4 mutations, specifically in RPE1 and HEK293T cell lines. Using a combination of CRISPR/Cas9 and lentiviral transduction technologies, both myc-tagged wild-type and mutant (G516R and R729W) COG4 proteins were expressed under the endogenous COG4 promoter. Constructed isogenic cell lines were comprehensively characterized using biochemical, microscopy (superresolution and electron), and proteomics approaches. The analysis revealed similar stability and localization of COG complex subunits, wild-type cell growth, and normal Golgi morphology in all three cell lines. Importantly, COG4-G516R cells demonstrated increased HPA-647 binding to the plasma membrane glycoconjugates, while COG4-R729W cells revealed high GNL-647 binding, indicating specific defects in O- and N-glycosylation. Both mutant cell lines express an elevated level of heparin sulfate proteoglycans. Moreover, a quantitative mass-spectrometry analysis of proteins secreted by COG-deficient cell lines revealed abnormal secretion of SIL1 and ERGIC-53 proteins by COG4-G516R cells. Interestingly, the clinical phenotype of patients with congenital mutations in the SIL1 gene (Marinesco-Sjogren syndrome) overlaps with the phenotype of COG4-G516R patients (Saul-Wilson syndrome). Our work is the first compressive study involving the creation of different COG mutations in different cell lines other than the patient's fibroblast. It may help to address the underlying cause of the phenotypic defects leading to the discovery of a proper treatment guideline for COG-CDGs.
Collapse
Affiliation(s)
| | | | - Vladimir Lupashin
- Department of Physiology and Cell Biology, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| |
Collapse
|
4
|
Lemarié M, Chatonnet F, Caron G, Fest T. Early Emergence of Adaptive Mechanisms Sustaining Ig Production: Application to Antibody Therapy. Front Immunol 2021; 12:671998. [PMID: 33995412 PMCID: PMC8117215 DOI: 10.3389/fimmu.2021.671998] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Accepted: 04/12/2021] [Indexed: 01/13/2023] Open
Abstract
Antibody therapy, where artificially-produced immunoglobulins (Ig) are used to treat pathological conditions such as auto-immune diseases and cancers, is a very innovative and competitive field. Although substantial efforts have been made in recent years to obtain specific and efficient antibodies, there is still room for improvement especially when considering a precise tissular targeting or increasing antigen affinity. A better understanding of the cellular and molecular steps of terminal B cell differentiation, in which an antigen-activated B cell becomes an antibody secreting cell, may improve antibody therapy. In this review, we use our recently published data about human B cell differentiation, to show that the mechanisms necessary to adapt a metamorphosing B cell to its new secretory function appear quite early in the differentiation process i.e., at the pre-plasmablast stage. After characterizing the molecular pathways appearing at this stage, we will focus on recent findings about two main processes involved in antibody production: unfolded protein response (UPR) and endoplasmic reticulum (ER) stress. We’ll show that many genes coding for factors involved in UPR and ER stress are induced at the pre-plasmablast stage, sustaining our hypothesis. Finally, we propose to use this recently acquired knowledge to improve productivity of industrialized therapeutic antibodies.
Collapse
Affiliation(s)
- Maud Lemarié
- Université de Rennes 1, INSERM, Établissement Français du Sang de Bretagne, UMR_S1236, Rennes, France
| | - Fabrice Chatonnet
- Université de Rennes 1, INSERM, Établissement Français du Sang de Bretagne, UMR_S1236, Rennes, France.,Laboratoire d'Hématologie, Pôle de Biologie, Centre Hospitalier Universitaire, Rennes, France
| | - Gersende Caron
- Université de Rennes 1, INSERM, Établissement Français du Sang de Bretagne, UMR_S1236, Rennes, France.,Laboratoire d'Hématologie, Pôle de Biologie, Centre Hospitalier Universitaire, Rennes, France
| | - Thierry Fest
- Université de Rennes 1, INSERM, Établissement Français du Sang de Bretagne, UMR_S1236, Rennes, France.,Laboratoire d'Hématologie, Pôle de Biologie, Centre Hospitalier Universitaire, Rennes, France
| |
Collapse
|
5
|
Ichhaporia VP, Hendershot LM. Role of the HSP70 Co-Chaperone SIL1 in Health and Disease. Int J Mol Sci 2021; 22:ijms22041564. [PMID: 33557244 PMCID: PMC7913895 DOI: 10.3390/ijms22041564] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 01/30/2021] [Accepted: 01/30/2021] [Indexed: 12/04/2022] Open
Abstract
Cell surface and secreted proteins provide essential functions for multicellular life. They enter the endoplasmic reticulum (ER) lumen co-translationally, where they mature and fold into their complex three-dimensional structures. The ER is populated with a host of molecular chaperones, associated co-factors, and enzymes that assist and stabilize folded states. Together, they ensure that nascent proteins mature properly or, if this process fails, target them for degradation. BiP, the ER HSP70 chaperone, interacts with unfolded client proteins in a nucleotide-dependent manner, which is tightly regulated by eight DnaJ-type proteins and two nucleotide exchange factors (NEFs), SIL1 and GRP170. Loss of SIL1′s function is the leading cause of Marinesco-Sjögren syndrome (MSS), an autosomal recessive, multisystem disorder. The development of animal models has provided insights into SIL1′s functions and MSS-associated pathologies. This review provides an in-depth update on the current understanding of the molecular mechanisms underlying SIL1′s NEF activity and its role in maintaining ER homeostasis and normal physiology. A precise understanding of the underlying molecular mechanisms associated with the loss of SIL1 may allow for the development of new pharmacological approaches to treat MSS.
Collapse
|
6
|
Chiesa R, Sallese M. Review: Protein misfolding diseases – the rare case of Marinesco‐Sjögren syndrome. Neuropathol Appl Neurobiol 2020; 46:323-343. [DOI: 10.1111/nan.12588] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Accepted: 11/03/2019] [Indexed: 12/15/2022]
Affiliation(s)
- R. Chiesa
- Department of Neuroscience Istituto di Ricerche Farmacologiche Mario Negri IRCCS MilanItaly
| | - M. Sallese
- Department of Medical, Oral and Biotechnological Sciences University "G. d'Annunzio" Chieti Italy
- CeSI‐MeT Center for Research on Ageing and Translational Medicine University "G. d'Annunzio" Chieti Italy
| |
Collapse
|
7
|
Salimi L, Salarinasab S, Rahbarghazi R, Nourazarian A, Nikanfar M, Avci ÇB, Bagca BG, Ozates Ay NP, Hasanpour M. High Glucose Content Abrogated the Normal Activity of Heat Shock Protein Signaling Pathway in Human Neuroblastoma Cells. Arch Med Res 2020; 51:180-184. [PMID: 32111494 DOI: 10.1016/j.arcmed.2020.01.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Revised: 12/18/2019] [Accepted: 01/20/2020] [Indexed: 11/25/2022]
Abstract
BACKGROUND Detrimental effects of high glucose content (HGC) were proved in different tissues such as the central nervous system. It seems that diabetic conditions could also alter the functional behavior of stem cells residing in the context of the nervous system. METHODS The possible effects of 40 and 70 mmol glucose were examined on HSP70 signaling pathways with a specific focus on protein translation, folding values of human neuroblastoma cell line SHSY-5Y after 72 h. Human neuroblastoma cells were exposed to 5, 40 and 70 mmol glucose doses. The transcription level of genes related to HSP70 signaling was also evaluated by PCR array. RESULTS The data from PCR array showed high glucose especially 70 mmol could potentially modulate the normal function of protein folding, endoplasmic reticulum derived protein folding and synthesis in neuroblastoma cells (p <0.05). CONCLUSIONS Data showed that high glucose condition makes neuroblastoma cells prone to biochemical insufficiency by affecting the function of HSP70 signaling pathway and protein synthesis.
Collapse
Affiliation(s)
- Leila Salimi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sadegh Salarinasab
- Neuroscience Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| | - Reza Rahbarghazi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Applied Cell Sciences, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Alireza Nourazarian
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Biochemistry and Clinical Laboratories, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Masoud Nikanfar
- Department of Neurology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Çıgır Biray Avci
- Department of Medical Biology, Faculty of Medicine, Ege University, Izmir, Turkey
| | - Bakiye Goker Bagca
- Department of Medical Biology, Faculty of Medicine, Ege University, Izmir, Turkey
| | | | - Milad Hasanpour
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Biochemistry and Clinical Laboratories, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
8
|
Functional Role of SIL1 in Neurodevelopment and Learning. Neural Plast 2019; 2019:9653024. [PMID: 31531014 PMCID: PMC6720716 DOI: 10.1155/2019/9653024] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Revised: 06/02/2019] [Accepted: 07/01/2019] [Indexed: 11/17/2022] Open
Abstract
Background Sil1 is the causative gene of Marinesco-Sjӧgren Syndrome (MSS). The mutated Sil1 generates shortened SIL1 protein which will form aggregation and be degraded rapidly. Mental retardation is a major symptom of MSS which suggests a role of SIL1 in the development of the central nervous system, but how SIL1 functions remains unclear. Objectives The aim of this study is to explore the role of SIL1 in regulating cerebral development and its underlying molecular mechanism. Methods The basic expression pattern of SIL1 in tissues and cultured cortical neurons is measured by immunostaining and Western blot. The expression of SIL1 is reduced in vitro and in vivo through RNA interference delivered by a lentivirus. The expression of NMDA receptor subunits and the function of the Reelin signaling pathway are then examined by surface biotinylation and Western blot subsequently. Finally, the spatial learning of young mice was assessed by the Barnes maze task. Results SIL1 deficiency caused a diminished expression of both Reelin receptors and therefore impaired the Reelin signaling pathway. It then inhibited the developmental expression of GluN2A and impaired the spatial learning of 5-week-old mice. Conclusions These results suggested that SIL1 is required for the development of the central nervous system which is associated with its role in Reelin signaling.
Collapse
|
9
|
Capone V, Clemente E, Restelli E, Di Campli A, Sperduti S, Ornaghi F, Pietrangelo L, Protasi F, Chiesa R, Sallese M. PERK inhibition attenuates the abnormalities of the secretory pathway and the increased apoptotic rate induced by SIL1 knockdown in HeLa cells. Biochim Biophys Acta Mol Basis Dis 2018; 1864:3164-3180. [DOI: 10.1016/j.bbadis.2018.07.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Revised: 06/05/2018] [Accepted: 07/03/2018] [Indexed: 02/06/2023]
|
10
|
Xu H, Xu S, Zhang R, Xin T, Pang Q. SIL1 functions as an oncogene in glioma by AKT/mTOR signaling pathway. Onco Targets Ther 2018; 11:3775-3783. [PMID: 29997438 PMCID: PMC6033116 DOI: 10.2147/ott.s167552] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Purpose SIL1 is a ubiquitous protein localized to the endoplasmic reticulum and functions as a cochaperone of BiP. Previous studies have shown that function loss of SIL1 is often associated with neurological diseases, such as Marinesco-Sjögren Syndrome. However, no studies have investigated the function of SIL1 in tumors. In this study we aim to reveal functions of SIL1 and the underlying mechanisms in glioma. Materials and methods First, by searching on Gene Expression Profiling Interactive Analysis, we examined SIL1 expression and prognostic value in glioblastoma multiforme (GBM) and brain lower grade glioma (LGG). Immunohistochemical analysis (IHC) was also performed to determine the endogenic SIL1 level. Cell counting kit-8 (CCK8) and clone formation assays were used to detect cell proliferation of U251 cells. Cell migration was detected by transwell assay and cell cycle and apoptosis were detected by flow cytometry. Western blot was performed to determine protein expression. Results We found that the expression of SIL1 was increased by approximately 1.5-fold in GBM and 1.3-fold in LGG compared with normal controls (P<0.05) and negatively correlated with patients’ survival. IHC revealed that SIL1 expression was significantly higher in glioma tissues than that in paracancerous tissues (P<0.05). Glioma patients with high SIL1 expression accounted for 65.79% (25/38) of total samples and SIL1 expression significantly increased in grade IV glioma compared to grades I–III (P=0.026). Suppression of SIL1 expression led to significant inhibition of U251 cell proliferation. Transwell assay showed that cell migration of U251 was significantly inhibited by siSIL transfection, with an inhibitory rate reaching 69%. Flow cytometry detection showed that siSIL1 could induce apoptosis of U251 cells and upregulated the expression of the pro-apoptotic protein Bax and Caspase3-P17. However, siSIL1 transfection had no effect on the cell cycle. Mechanism studies demonstrated that siSIL1 transfection led to inactivation of AKT/mTOR signaling pathway, including decreased phosphorylation of AKT and mTOR without affecting protein expression, as well as decreased expression of the downstream effector p70S6K. Conclusion Downregulation of SIL1 inhibited the progression of glioma by suppressing the AKT/mTOR signaling pathway.
Collapse
Affiliation(s)
- Hao Xu
- Department of Neurosurgery, Shandong Provincial Hospital Affiliated to Shandong University, Jinan 250021, Shandong, China, ;
| | - Shangchen Xu
- Department of Neurosurgery, Shandong Provincial Hospital Affiliated to Shandong University, Jinan 250021, Shandong, China, ;
| | - Rui Zhang
- Department of Neurosurgery, Shandong Provincial Hospital Affiliated to Shandong University, Jinan 250021, Shandong, China, ;
| | - Tao Xin
- Department of Neurosurgery, Shandong Provincial Hospital Affiliated to Shandong University, Jinan 250021, Shandong, China, ;
| | - Qi Pang
- Department of Neurosurgery, Shandong Provincial Hospital Affiliated to Shandong University, Jinan 250021, Shandong, China, ;
| |
Collapse
|
11
|
Ichhaporia VP, Kim J, Kavdia K, Vogel P, Horner L, Frase S, Hendershot LM. SIL1, the endoplasmic-reticulum-localized BiP co-chaperone, plays a crucial role in maintaining skeletal muscle proteostasis and physiology. Dis Model Mech 2018; 11:dmm.033043. [PMID: 29666155 PMCID: PMC5992605 DOI: 10.1242/dmm.033043] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2017] [Accepted: 03/22/2018] [Indexed: 01/02/2023] Open
Abstract
Mutations in SIL1, a cofactor for the endoplasmic reticulum (ER)-localized Hsp70 chaperone, BiP, cause Marinesco-Sjögren syndrome (MSS), an autosomal recessive disorder. Using a mouse model, we characterized molecular aspects of the progressive myopathy associated with MSS. Proteomic profiling of quadriceps at the onset of myopathy revealed that SIL1 deficiency affected multiple pathways critical to muscle physiology. We observed an increase in ER chaperones prior to the onset of muscle weakness, which was complemented by upregulation of multiple components of cellular protein degradation pathways. These responses were inadequate to maintain normal expression of secretory pathway proteins, including insulin and IGF-1 receptors. There was a paradoxical enhancement of downstream PI3K-AKT-mTOR signaling and glucose uptake in SIL1-disrupted skeletal muscles, all of which were insufficient to maintain skeletal muscle mass. Together, these data reveal a disruption in ER homeostasis upon SIL1 loss, which is countered by multiple compensatory responses that are ultimately unsuccessful, leading to trans-organellar proteostasis collapse and myopathy. Editor's choice: This study provides molecular insights into the progressive myopathy and cellular compensatory responses attempted upon loss of SIL1, a component of the endoplasmic-reticulum-resident Hsp70 protein-folding machinery.
Collapse
Affiliation(s)
- Viraj P Ichhaporia
- Dept of Microbiology, Immunology, and Biochemistry, The University of Tennessee Health Science Center, Memphis, TN 38163, USA.,Dept of Tumor Cell Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Jieun Kim
- Small Animal Imaging Center, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Kanisha Kavdia
- Proteomics Facility, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Peter Vogel
- Dept of Pathology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Linda Horner
- Cell and Tissue Imaging Center, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Sharon Frase
- Cell and Tissue Imaging Center, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Linda M Hendershot
- Dept of Microbiology, Immunology, and Biochemistry, The University of Tennessee Health Science Center, Memphis, TN 38163, USA .,Dept of Tumor Cell Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| |
Collapse
|
12
|
Buchkremer S, González Coraspe JA, Weis J, Roos A. Sil1-Mutant Mice Elucidate Chaperone Function in Neurological Disorders. J Neuromuscul Dis 2018; 3:169-181. [PMID: 27854219 PMCID: PMC5271578 DOI: 10.3233/jnd-160152] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Chaperone dysfunction leading to the build-up of misfolded proteins could frequently be linked to clinical manifestations also affecting the nervous system and the skeletal muscle. In addition, increase in chaperone function is beneficial to antagonize protein aggregation and thus represents a promising target for therapeutic concepts for many genetic and acquired chaperonopathies. However, little is known on the precise molecular mechanisms defining the cell and tissue abnormalities in the case of impaired chaperone function as well as on underlying effects in the case of compensatory up-regulation of chaperones. This scarcity of knowledge often arises from a lack of appropriate animal models that mimic closely the human molecular, cellular, and histological characteristics. Here, we introduce the Sil1-mutant woozy mouse as a suitable model to investigate molecular and cellular mechanisms of impaired ER-chaperone function affecting the integrity of nervous system and skeletal muscle. The overlapping clinical findings in man and mouse indicate that woozy is a good copy of a human phenotype called Marinesco-Sjögren syndrome. We confirm the presence of ER-stress and expand the biochemical knowledge of altered nuclear envelope in muscle, a hallmark of SIL1-disease. In addition, our data suggest that impaired excitation-contraction coupling might be part of the SIL1-pathophysiology. Our results moreover indicate that divergent expression of pro- and anti-survival proteins is decisive for Purkinje cell survival. By summarizing the current knowledge of woozy, we focus on the suitability of this animal model to study neuroprotective co-chaperone function and to investigate the involvement of co-chaperones in the predisposition of other disorders such as diabetic neuropathy.
Collapse
Affiliation(s)
- Stephan Buchkremer
- Institute of Neuropathology, University Hospital RWTH Aachen, Aachen, Germany
| | | | - Joachim Weis
- Institute of Neuropathology, University Hospital RWTH Aachen, Aachen, Germany
| | - Andreas Roos
- Institute of Neuropathology, University Hospital RWTH Aachen, Aachen, Germany.,Leibniz-Institut für Analytische Wissenschaften ISAS e.V., Dortmund, Germany.,The John Walton Muscular Dystrophy Research Centre, MRC Centre for Neuromuscular Diseases, Newcastle University, Central Parkway, Newcastle upon Tyne, UK
| |
Collapse
|
13
|
Targeting the enhanced ER stress response in Marinesco-Sjögren syndrome. J Neurol Sci 2017; 385:49-56. [PMID: 29406913 DOI: 10.1016/j.jns.2017.12.010] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2017] [Revised: 11/11/2017] [Accepted: 12/08/2017] [Indexed: 12/23/2022]
Abstract
BACKGROUND AND OBJECTIVE Marinesco-Sjögren syndrome (MSS) is an autosomal recessive infantile-onset disorder characterized by cataracts, cerebellar ataxia, and progressive myopathy caused by mutation of SIL1. In mice, a defect in SIL1 causes endoplasmic reticulum (ER) chaperone dysfunction, leading to unfolded protein accumulation and increased ER stress. However, ER stress and the unfolded protein response (UPR) have not been investigated in MSS patient-derived cells. METHODS Lymphoblastoid cell lines (LCLs) were established from four MSS patients. Spontaneous and tunicamycin-induced ER stress and the UPR were investigated in MSS-LCLs. Expression of UPR markers was analyzed by western blotting. ER stress-induced apoptosis was analyzed by flow cytometry. The cytoprotective effects of ER stress modulators were also examined. RESULTS MSS-LCLs exhibited increased spontaneous ER stress and were highly susceptible to ER stress-induced apoptosis. The inositol-requiring protein 1α (IRE1α)-X-box-binding protein 1 (XBP1) pathway was mainly upregulated in MSS-LCLs. Tauroursodeoxycholic acid (TUDCA) attenuated ER stress-induced apoptosis. CONCLUSION MSS patient-derived cells exhibit increased ER stress, an activated UPR, and susceptibility to ER stress-induced death. TUDCA reduces ER stress-induced death of MSS patient-derived cells. The potential of TUDCA as a therapeutic agent for MSS could be explored further in preclinical studies.
Collapse
|
14
|
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: 14] [Impact Index Per Article: 2.0] [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
|
15
|
Jin H, Komita M, Aoe T. The Role of BiP Retrieval by the KDEL Receptor in the Early Secretory Pathway and its Effect on Protein Quality Control and Neurodegeneration. Front Mol Neurosci 2017; 10:222. [PMID: 28769758 PMCID: PMC5511815 DOI: 10.3389/fnmol.2017.00222] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2017] [Accepted: 06/27/2017] [Indexed: 12/14/2022] Open
Abstract
Protein quality control in the early secretory pathway is a ubiquitous eukaryotic mechanism for adaptation to endoplasmic reticulum (ER) stress. An ER molecular chaperone, immunoglobulin heavy chain-binding protein (BiP), is one of the essential components in this process. BiP interacts with nascent proteins to facilitate their folding. BiP also plays an important role in preventing aggregation of misfolded proteins and regulating the ER stress response when cells suffer various injuries. BiP is a member of the 70-kDa heat shock protein (HSP70) family of molecular chaperones that resides in the ER. Interaction between BiP and unfolded proteins is mediated by a substrate-binding domain and a nucleotide-binding domain for ATPase activity, leading to protein folding and maturation. BiP also possesses a retrieval motif in its carboxyl terminal. When BiP is secreted from the ER, the Lys-Asp-Glu-Leu (KDEL) receptor in the post-ER compartments binds with the carboxyl terminal KDEL sequence of BiP and returns BiP to the ER via coat protein complex I (COPI) vesicular transport. Although yeast studies showed that BiP retrieval by the KDEL receptor is not essential in single cells, it is crucial for multicellular organisms, where some essential proteins require retrieval to facilitate folding and maturation. Experiments in knock-in mice expressing mutant BiP with the retrieval motif deleted revealed a unique role of BiP retrieval by the KDEL receptor in neuronal development and age-related neurodegeneration.
Collapse
Affiliation(s)
- Hisayo Jin
- Department of Anesthesiology, Graduate School of Medicine, Chiba UniversityChiba, Japan
| | - Mari Komita
- Department of Anesthesiology, Chiba Rosai HospitalIchihara, Japan
| | - Tomohiko Aoe
- Pain Center, Chiba Medical Center, Teikyo UniversityIchihara, Japan
| |
Collapse
|
16
|
Behnke J, Feige MJ, Hendershot LM. BiP and its nucleotide exchange factors Grp170 and Sil1: mechanisms of action and biological functions. J Mol Biol 2015; 427:1589-608. [PMID: 25698114 DOI: 10.1016/j.jmb.2015.02.011] [Citation(s) in RCA: 135] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2014] [Revised: 02/10/2015] [Accepted: 02/10/2015] [Indexed: 12/26/2022]
Abstract
BiP (immunoglobulin heavy-chain binding protein) is the endoplasmic reticulum (ER) orthologue of the Hsp70 family of molecular chaperones and is intricately involved in most functions of this organelle through its interactions with a variety of substrates and regulatory proteins. Like all Hsp70 family members, the ability of BiP to bind and release unfolded proteins is tightly regulated by a cycle of ATP binding, hydrolysis, and nucleotide exchange. As a characteristic of the Hsp70 family, multiple DnaJ-like co-factors can target substrates to BiP and stimulate its ATPase activity to stabilize the binding of BiP to substrates. However, only in the past decade have nucleotide exchange factors for BiP been identified, which has shed light not only on the mechanism of BiP-assisted folding in the ER but also on Hsp70 family members that reside throughout the cell. We will review the current understanding of the ATPase cycle of BiP in the unique environment of the ER and how it is regulated by the nucleotide exchange factors, Grp170 (glucose-regulated protein of 170kDa) and Sil1, both of which perform unanticipated roles in various biological functions and disease states.
Collapse
Affiliation(s)
- Julia Behnke
- Department of Tumor Cell Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Matthias J Feige
- Department of Tumor Cell Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Linda M Hendershot
- Department of Tumor Cell Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA.
| |
Collapse
|