1
|
Tan L, He G, Shen C, He S, Chen Y, Guo X. Construction of a ferroptosis-based prediction model for the prognosis of MYCN-amplified neuroblastoma and screening and verification of target sites. Hereditas 2025; 162:41. [PMID: 40108662 PMCID: PMC11921587 DOI: 10.1186/s41065-025-00413-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2025] [Accepted: 03/09/2025] [Indexed: 03/22/2025] Open
Abstract
BACKGROUND Neuroblastoma (NB) is a prevalent extracranial solid tumor in pediatric patients. Of these, the MYCN-amplified type has a poor treatment response and prognosis. To enhance therapeutic efficacy and prognostic outcomes, numerous research teams have undertaken extensive investigations through various pathways and directions. Among these, ferroptosis has recently emerged as a significant area of research focus.Ferroptosis, a type of iron-dependent cell death, is primarily caused by lipid peroxides. This study intends to develop a prognosis model based on MYCN-amplified NB and ferroptosis-related genes (FGs). METHODS Data for this study were sourced from the TARGET and FerrDb databases. Lasso regression algorithms and univariate COX analysis were leveraged to determine feature genes; multivariate COX analysis was employed to develop a prediction model and risk scores; and receiver operating characteristic (ROC) curves and Kaplan-Meier analysis were utilized to assess the predictive ability of the model. Furthermore, discrepancies in immune cell infiltration (ICI) between the high-risk (HR) and low-risk (LR) populations were assessed via CIBERSORT analysis. Finally, experiments were conducted on MYCN-amplified and MYCN non-amplified cells so as to validate the differential expression of the gene. RESULTS A prediction model was constructed and risk scores were calculated based on 4 genes (LIFR, TP53, NRAS, and OSBPL9). The HR group, which was stratified by the median score, had a lower overall survival rate than the LR group.The differences in expression of each gene between MYCN-amplified and MYCN non-amplified cells were further confirmed through cell experiments and qPCR. CONCLUSION The prediction model in this study can be employed to forecast the prognosis of MYCN-amplified NB. These genes may represent promising new ferroptosis-related intervention targets (FITs) in treating MYCN-amplified NB, with the potential to improve patient outcomes.
Collapse
Affiliation(s)
- Linjun Tan
- Department of Pediatrics, Affiliated Hospital of Zunyi Medical University, No.149 Dalian Road, Huichuan District, Zunyi, 563003, Guizhou, China
- Department of Pediatrics, Guizhou Children's Hospital, No.149 Dalian Road, Huichuan District, Zunyi, 563003, Guizhou, China
| | - Guoqian He
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Section 3, South Renmin Road, Chengdu, 610041, China
| | - Chengqi Shen
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Section 3, South Renmin Road, Chengdu, 610041, China
| | - Sijia He
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Section 3, South Renmin Road, Chengdu, 610041, China
| | - Yan Chen
- Department of Pediatrics, Affiliated Hospital of Zunyi Medical University, No.149 Dalian Road, Huichuan District, Zunyi, 563003, Guizhou, China.
- Department of Pediatrics, Guizhou Children's Hospital, No.149 Dalian Road, Huichuan District, Zunyi, 563003, Guizhou, China.
- Collaborative Innovation Center for Tissue Injury Repairand Regenerative Medicine of Zunyi Medical University, No.149 Dalian Road, Huichuan District, Zunyi, 563003, Guizhou, China.
| | - Xia Guo
- Department of Pediatrics, Affiliated Hospital of Zunyi Medical University, No.149 Dalian Road, Huichuan District, Zunyi, 563003, Guizhou, China.
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Section 3, South Renmin Road, Chengdu, 610041, China.
| |
Collapse
|
2
|
Liu L, Han F, Du N, Liu Y, Duan A, Kang S, Li B. New insights into the ferroptosis and immune infiltration in endometriosis: a bioinformatics-based analysis. Front Immunol 2025; 15:1507083. [PMID: 39872538 PMCID: PMC11769811 DOI: 10.3389/fimmu.2024.1507083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Accepted: 12/26/2024] [Indexed: 01/30/2025] Open
Abstract
Background Ferroptosis, a recently discovered iron-dependent cell death, is linked to various diseases but its role in endometriosis is still not fully understood. Methods In this study, we integrated microarray data of endometriosis from the GEO database and ferroptosis-related genes (FRGs) from the FerrDb database to further investigate the regulation of ferroptosis in endometriosis and its impact on the immune microenvironment. WGCNA identified ferroptosis-related modules, annotated by GO & KEGG. MNC algorithm pinpointed hub FRGs. Cytoscape construct a ceRNA network, and ROC curves evaluated diagnostic efficacy of hub FRGs. Consensus cluster analysis identified ferroptosis subclusters, and CIBERSORT assessed immune infiltration of these subclusters. Finally, RT-qPCR validated hub FRG expression in clinical tissues. Results We identified two ferroptosis modules of endometriosis, and by enrichment analysis, they are closely linked with autophagy, mTOR, oxidative stress, and FOXO pathways. Furthermore, we identified 10 hub FRGs, and the ROC curve showed better predictive ability for diagnosing. RT-qPCR confirmed that the tissue expression of 10 hub FRGs was mostly consistent with the database results. Subsequently, we developed a ceRNA network based on 4 FRGs (BECN1, OSBPL9, TGFBR1, GSK3B). Next, we identified two ferroptosis subclusters of endometriosis and discovered that they are closely linked with endometriosis stage. Importantly, immune enrichment analysis illustrated that the expression levels of immune cells and immune checkpoint genes were significantly different in the two ferroptosis subclusters. Specifically, the ferroptosis subcluster with stage III-IV of endometriosis is more inclined to the immunosuppressive microenvironment. Conclusions Our study showed that ferroptosis may jointly promote endometriosis progression by remodeling the immune microenvironment, offering new insights into pathogenesis and therapeutics.
Collapse
Affiliation(s)
- Lusha Liu
- Department of Gynecology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Feifei Han
- Department of Gynecology, Handan Central Hospital, Handan, China
| | - Naiyi Du
- Department of Gynecology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Yakun Liu
- Department of Gynecology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Aihong Duan
- Department of Gynecology, Handan Central Hospital, Handan, China
| | - Shan Kang
- Department of Gynecology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Bin Li
- Department of Gynecology, Handan Central Hospital, Handan, China
| |
Collapse
|
3
|
Kawasaki A, Nakatsu F. Measurement of ORP10-Mediated Lipid Countertransport at ER-Endosome Membrane Contact Sites via a Chemically Induced Dimerization Strategy. Methods Mol Biol 2025; 2888:13-22. [PMID: 39699721 DOI: 10.1007/978-1-0716-4318-1_2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2024]
Abstract
Oxysterol-binding protein (OSBP)-related proteins (ORPs) are a large family of lipid transfer proteins (LTPs) in mammals. ORPs mediate the countertransport of two distinct lipids at membrane contact sites (MCSs). ORP10 is localized via binding to ORP9 at the endoplasmic reticulum (ER)-endosome MCSs, where it mediates countertransport of phosphatidylinositol 4-phosphate (PI4P) and phosphatidylserine (PS). To quantitatively monitor the lipid countertransport process mediated by ORP10 in situ, we take advantage of chemically induced dimerization (CID), a strategy of inducing protein-protein interactions by exposure to chemicals. Specifically, we exploit the rapamycin-inducible heterodimerization of FKBP/FRB to acutely recruit the lipid transfer domain of ORP10 to the ER-endosome MCSs and monitor the levels of PI4P and PS on endosomes by their genetic probes in live imaging. This approach enables the measurement of ORP10 activity in lipid countertransport at ER-endosome MCSs and is also beneficial as a versatile method applicable to other LTPs.
Collapse
Affiliation(s)
- Asami Kawasaki
- Department of Neurochemistry and Molecular Cell Biology, Niigata University School of Medicine and Graduate School of Medical/Dental Sciences, Niigata, Japan
| | - Fubito Nakatsu
- Department of Neurochemistry and Molecular Cell Biology, Niigata University School of Medicine and Graduate School of Medical/Dental Sciences, Niigata, Japan.
| |
Collapse
|
4
|
Lin S, Chang P, Tsao S, Aderinwale A, Sallapalli BT, He J, Zhang Y. Oxysterol binding protein (OSBP) contributes to hepatitis E virus replication. Virol J 2024; 21:161. [PMID: 39039546 PMCID: PMC11265327 DOI: 10.1186/s12985-024-02438-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Accepted: 07/16/2024] [Indexed: 07/24/2024] Open
Abstract
Hepatitis E virus (HEV) is a positive-sense, single-stranded RNA virus and causes primarily acute self-limiting infections. The ORF1 of the HEV genome encodes a polyprotein around 190 kDa, which contains several putative domains, including helicase and RNA-dependent RNA polymerase. The HEV-encoded helicase is a member of the superfamily 1 helicase family and possesses multiple enzymatic functions, such as RNA 5'-triphosphatase, RNA unwinding, and NTPase, which are thought to contribute to viral RNA synthesis. However, the helicase interaction with cellular proteins remains less known. Oxysterol binding protein (OSBP) is a lipid regulator that shuffles between the Golgi apparatus and the endoplasmic reticulum for cholesterol and phosphatidylinositol-4-phosphate exchange and controls the efflux of cholesterol from cells. In this study, the RNAi-mediated silencing of OSBP significantly reduced HEV replication. Further studies indicate that the HEV helicase interacted with OSBP, shown by co-immunoprecipitation and co-localization in co-transfected cells. The presence of helicase blocked OSBP preferential translocation to the Golgi apparatus. These results demonstrate that OSBP contributes to HEV replication and enrich our understanding of the HEV-cell interactions.
Collapse
Affiliation(s)
- Shaoli Lin
- Molecular Virology Laboratory, VA-MD College of Veterinary Medicine, University of Maryland, College Park, MD, USA
| | - Peixi Chang
- Molecular Virology Laboratory, VA-MD College of Veterinary Medicine, University of Maryland, College Park, MD, USA
| | - Shane Tsao
- National Cancer Institute, National Institute of Health, Bethesda, MD, USA
| | - Abigail Aderinwale
- Molecular Virology Laboratory, VA-MD College of Veterinary Medicine, University of Maryland, College Park, MD, USA
| | - Bhargava Teja Sallapalli
- Molecular Virology Laboratory, VA-MD College of Veterinary Medicine, University of Maryland, College Park, MD, USA
| | - Jia He
- Molecular Virology Laboratory, VA-MD College of Veterinary Medicine, University of Maryland, College Park, MD, USA
| | - Yanjin Zhang
- Molecular Virology Laboratory, VA-MD College of Veterinary Medicine, University of Maryland, College Park, MD, USA.
| |
Collapse
|
5
|
Monteiro-Cardoso VF, Giordano F. Emerging functions of the mitochondria-ER-lipid droplet three-way junction in coordinating lipid transfer, metabolism, and storage in cells. FEBS Lett 2024; 598:1252-1273. [PMID: 38774950 DOI: 10.1002/1873-3468.14893] [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: 02/05/2024] [Revised: 03/18/2024] [Accepted: 04/05/2024] [Indexed: 05/25/2024]
Abstract
Over the past two decades, we have witnessed a growing appreciation for the importance of membrane contact sites (CS) in facilitating direct communication between organelles. CS are tiny regions where the membranes of two organelles meet but do not fuse and allow the transfer of metabolites between organelles, playing crucial roles in the coordination of cellular metabolic activities. The significant advancements in imaging techniques and molecular and cell biology research have revealed that CS are more complex than what originally thought, and as they are extremely dynamic, they can remodel their shape, composition, and functions in accordance with metabolic and environmental changes and can occur between more than two organelles. Here, we describe how recent studies led to the identification of a three-way mitochondria-ER-lipid droplet CS and discuss the emerging functions of these contacts in maintaining lipid storage, homeostasis, and balance. We also summarize the properties and functions of key protein components localized at the mitochondria-ER-lipid droplet interface, with a special focus on lipid transfer proteins. Understanding tripartite CS is essential for unraveling the complexities of inter-organelle communication and cooperation within cells.
Collapse
Affiliation(s)
- Vera Filipa Monteiro-Cardoso
- Institute for Integrative Biology of the Cell (I2BC), CEA, CNRS, Université Paris-Saclay, Gif-sur-Yvette cedex, France
- Inserm U1280, Gif-sur-Yvette cedex, France
| | - Francesca Giordano
- Institute for Integrative Biology of the Cell (I2BC), CEA, CNRS, Université Paris-Saclay, Gif-sur-Yvette cedex, France
- Inserm U1280, Gif-sur-Yvette cedex, France
| |
Collapse
|
6
|
Mavuduru VA, Vadupu L, Ghosh KK, Chakrabortty S, Gulyás B, Padmanabhan P, Ball WB. Mitochondrial phospholipid transport: Role of contact sites and lipid transport proteins. Prog Lipid Res 2024; 94:101268. [PMID: 38195013 DOI: 10.1016/j.plipres.2024.101268] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Revised: 01/03/2024] [Accepted: 01/05/2024] [Indexed: 01/11/2024]
Abstract
One of the major constituents of mitochondrial membranes is the phospholipids, which play a key role in maintaining the structure and the functions of the mitochondria. However, mitochondria do not synthesize most of the phospholipids in situ, necessitating the presence of phospholipid import pathways. Even for the phospholipids, which are synthesized within the inner mitochondrial membrane (IMM), the phospholipid precursors must be imported from outside the mitochondria. Therefore, the mitochondria heavily rely on the phospholipid transport pathways for its proper functioning. Since, mitochondria are not part of a vesicular trafficking network, the molecular mechanisms of how mitochondria receive its phospholipids remain a relevant question. One of the major ways that hydrophobic phospholipids can cross the aqueous barrier of inter or intraorganellar spaces is by apposing membranes, thereby decreasing the distance of transport, or by being sequestered by lipid transport proteins (LTPs). Therefore, with the discovery of LTPs and membrane contact sites (MCSs), we are beginning to understand the molecular mechanisms of phospholipid transport pathways in the mitochondria. In this review, we will present a brief overview of the recent findings on the molecular architecture and the importance of the MCSs, both the intraorganellar and interorganellar contact sites, in facilitating the mitochondrial phospholipid transport. In addition, we will also discuss the role of LTPs for trafficking phospholipids through the intermembrane space (IMS) of the mitochondria. Mechanistic insights into different phospholipid transport pathways of mitochondria could be exploited to vary the composition of membrane phospholipids and gain a better understanding of their precise role in membrane homeostasis and mitochondrial bioenergetics.
Collapse
Affiliation(s)
- Vijay Aditya Mavuduru
- Department of Biological Sciences, School of Engineering and Sciences, SRM University AP Andhra Pradesh, Guntur, Andhra Pradesh 522240, India
| | - Lavanya Vadupu
- Department of Biological Sciences, School of Engineering and Sciences, SRM University AP Andhra Pradesh, Guntur, Andhra Pradesh 522240, India
| | - Krishna Kanta Ghosh
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, 59 Nanyang Drive, Singapore, 636921, Singapore
| | - Sabyasachi Chakrabortty
- Department of Chemistry, School of Engineering and Sciences, SRM University AP Andhra Pradesh, Guntur, Andhra Pradesh 522502, India
| | - Balázs Gulyás
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, 59 Nanyang Drive, Singapore, 636921, Singapore; Cognitive Neuroimaging Centre, Nanyang Technological University, Singapore, 59 Nanyang Drive, 636921, Singapore; Department of Clinical Neuroscience, Karolinska Institute, Stockholm 17176, Sweden
| | - Parasuraman Padmanabhan
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, 59 Nanyang Drive, Singapore, 636921, Singapore; Cognitive Neuroimaging Centre, Nanyang Technological University, Singapore, 59 Nanyang Drive, 636921, Singapore.
| | - Writoban Basu Ball
- Department of Biological Sciences, School of Engineering and Sciences, SRM University AP Andhra Pradesh, Guntur, Andhra Pradesh 522240, India.
| |
Collapse
|
7
|
Criado Santos N, Bouvet S, Cruz Cobo M, Mandavit M, Bermont F, Castelbou C, Mansour F, Azam M, Giordano F, Nunes-Hasler P. Sec22b regulates phagosome maturation by promoting ORP8-mediated lipid exchange at endoplasmic reticulum-phagosome contact sites. Commun Biol 2023; 6:1008. [PMID: 37794132 PMCID: PMC10550925 DOI: 10.1038/s42003-023-05382-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Accepted: 09/21/2023] [Indexed: 10/06/2023] Open
Abstract
Phagosome maturation is critical for immune defense, defining whether ingested material is destroyed or converted into antigens. Sec22b regulates phagosome maturation, yet how has remained unclear. Here we show Sec22b tethers endoplasmic reticulum-phagosome membrane contact sites (MCS) independently of the known tether STIM1. Sec22b knockdown increases calcium signaling, phagolysosome fusion and antigen degradation and alters phagosomal phospholipids PI(3)P, PS and PI(4)P. Levels of PI(4)P, a lysosome docking lipid, are rescued by Sec22b re-expression and by expression of the artificial tether MAPPER but not the MCS-disrupting mutant Sec22b-P33. Moreover, Sec22b co-precipitates with the PS/PI(4)P exchange protein ORP8. Wild-type, but not mutant ORP8 rescues phagosomal PI(4)P and reduces antigen degradation. Sec22b, MAPPER and ORP8 but not P33 or mutant-ORP8 restores phagolysosome fusion in knockdown cells. These findings clarify an alternative mechanism through which Sec22b controls phagosome maturation and beg a reassessment of the relative contribution of Sec22b-mediated fusion versus tethering to phagosome biology.
Collapse
Affiliation(s)
- Nina Criado Santos
- Department of Pathology and Immunology, Geneva Center for Inflammation Research, Faculty of Medicine, University of Geneva, Centre Médicale Universitaire, 1 Rue Michel-Servet, Geneva, Switzerland
| | - Samuel Bouvet
- Department of Cellular Physiology and Metabolism, Faculty of Medicine, University of Geneva, Centre Médicale Universitaire, 1 Rue Michel-Servet, Geneva, Switzerland
| | - Maria Cruz Cobo
- Department of Pathology and Immunology, Geneva Center for Inflammation Research, Faculty of Medicine, University of Geneva, Centre Médicale Universitaire, 1 Rue Michel-Servet, Geneva, Switzerland
| | - Marion Mandavit
- Department of Pathology and Immunology, Geneva Center for Inflammation Research, Faculty of Medicine, University of Geneva, Centre Médicale Universitaire, 1 Rue Michel-Servet, Geneva, Switzerland
| | - Flavien Bermont
- Department of Cellular Physiology and Metabolism, Faculty of Medicine, University of Geneva, Centre Médicale Universitaire, 1 Rue Michel-Servet, Geneva, Switzerland
| | - Cyril Castelbou
- Department of Cellular Physiology and Metabolism, Faculty of Medicine, University of Geneva, Centre Médicale Universitaire, 1 Rue Michel-Servet, Geneva, Switzerland
| | - Farah Mansour
- Department of Pathology and Immunology, Geneva Center for Inflammation Research, Faculty of Medicine, University of Geneva, Centre Médicale Universitaire, 1 Rue Michel-Servet, Geneva, Switzerland
| | - Maral Azam
- Department of Pathology and Immunology, Geneva Center for Inflammation Research, Faculty of Medicine, University of Geneva, Centre Médicale Universitaire, 1 Rue Michel-Servet, Geneva, Switzerland
| | - Francesca Giordano
- Institute for Integrative Biology of the Cell (I2BC), CEA, CNRS, Université Paris-Saclay, Gif-sur-Yvette cedex, 91198, France
- Inserm U1280, Gif-sur-Yvette cedex, 91198, France
| | - Paula Nunes-Hasler
- Department of Pathology and Immunology, Geneva Center for Inflammation Research, Faculty of Medicine, University of Geneva, Centre Médicale Universitaire, 1 Rue Michel-Servet, Geneva, Switzerland.
| |
Collapse
|
8
|
Santos MF, Rappa G, Karbanová J, Diana P, Cirrincione G, Carbone D, Manna D, Aalam F, Wang D, Vanier C, Corbeil D, Lorico A. HIV-1-induced nuclear invaginations mediated by VAP-A, ORP3, and Rab7 complex explain infection of activated T cells. Nat Commun 2023; 14:4588. [PMID: 37563144 PMCID: PMC10415338 DOI: 10.1038/s41467-023-40227-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Accepted: 07/19/2023] [Indexed: 08/12/2023] Open
Abstract
The mechanism of human immunodeficiency virus 1 (HIV-1) nuclear entry, required for productive infection, is not fully understood. Here, we report that in HeLa cells and activated CD4+ T cells infected with HIV-1 pseudotyped with VSV-G and native Env protein, respectively, Rab7+ late endosomes containing endocytosed HIV-1 promote the formation of nuclear envelope invaginations (NEIs) by a molecular mechanism involving the VOR complex, composed of the outer nuclear membrane protein VAP-A, hyperphosphorylated ORP3 and Rab7. Silencing VAP-A or ORP3 and drug-mediated impairment of Rab7 binding to ORP3-VAP-A inhibited the nuclear transfer of the HIV-1 components and productive infection. In HIV-1-resistant quiescent CD4+ T cells, ORP3 was not hyperphosphorylated and neither VOR complex nor NEIs were formed. This new cellular pathway and its molecular players are potential therapeutic targets, perhaps shared by other viruses that require nuclear entry to complete their life cycle.
Collapse
Affiliation(s)
- Mark F Santos
- Touro University Nevada College of Osteopathic Medicine, Henderson, NV, USA
| | - Germana Rappa
- Touro University Nevada College of Osteopathic Medicine, Henderson, NV, USA
| | - Jana Karbanová
- Biotechnology Center (BIOTEC) and Center for Molecular and Cellular Bioengineering, Technische Universität Dresden, Dresden, Germany
| | - Patrizia Diana
- Department of Biological, Chemical, and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, Palermo, Italy
| | - Girolamo Cirrincione
- Department of Biological, Chemical, and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, Palermo, Italy
| | - Daniela Carbone
- Department of Biological, Chemical, and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, Palermo, Italy
| | - David Manna
- Touro College of Osteopathic Medicine, Middletown, New York, NY, USA
| | - Feryal Aalam
- Touro University Nevada College of Osteopathic Medicine, Henderson, NV, USA
| | - David Wang
- Touro University Nevada College of Osteopathic Medicine, Henderson, NV, USA
| | - Cheryl Vanier
- Touro University Nevada College of Osteopathic Medicine, Henderson, NV, USA
- Imgen Research, LLC, 5495 South Rainbow #201, Las Vegas, NV, USA
| | - Denis Corbeil
- Biotechnology Center (BIOTEC) and Center for Molecular and Cellular Bioengineering, Technische Universität Dresden, Dresden, Germany.
| | - Aurelio Lorico
- Touro University Nevada College of Osteopathic Medicine, Henderson, NV, USA.
| |
Collapse
|
9
|
Wang C, He Y, He Y, Liang W, Zhou C, Wu M, Meng Z, Li W, Cao J. Prognostic and biological function value of OSBPL3 in colorectal cancer analyzed by multi-omic data analysis. BMC Gastroenterol 2023; 23:270. [PMID: 37550605 PMCID: PMC10408063 DOI: 10.1186/s12876-023-02824-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Accepted: 05/17/2023] [Indexed: 08/09/2023] Open
Abstract
BACKGROUND Colorectal cancer (CRC) is one of the most common malignancies in the world. This study proposes to reveal prognostic biomarkers for the prognosis and treatment of CRC patients. METHODS Differential analysis of OSBPL3 was performed in pan-cancer, and the correlation between clinical stage and OSBPL3 was analyzed. Multiple omics analysis was used to compare the relationship between survival of patients and copy number variation, single nucleotide variant, and methylation status. Survival differences between high and low OSBPL3 expression groups were analyzed. Differentially expressed genes (DEGs) between high and low OSBPL3 expression groups were obtained, and functional enrichment analysis was implemented. Correlations between immune cells and OSBPL3 was analyzed. Drug sensitivity between the two OSBPL3 expression groups was compared. Moreover, the expression of OSBPL3 was verified by immunohistochemistry and real-time quantitative PCR. RESULTS OSBPL3 was differentially expressed in 13 tumors and had some correlations with T and N stages. OSBPL3 expression was regulated by methylation and higher OSBPL3 expression was associated with poorer prognosis in CRC. 128 DEGs were obtained and they were mainly involved in signaling receptor activator activity, aspartate and glutamate metabolism. T cell gamma delta and T cell follicular helper were significantly different in the high and low OSBPL3 expression groups. Moreover, OSBPL3 showed negative correlations with multiple drugs. OSBPL3 was significantly upregulated in CRC samples compared to normal samples. CONCLUSIONS A comprehensive analysis demonstrated that OSBPL3 had potential prognostic value, and guiding significance for CRC chemotherapeutic.
Collapse
Affiliation(s)
- Chengxing Wang
- The First Affiliated Hospital, Jinan University, Guangzhou, 529000, Guangdong, China
- Department of Gastrointestinal Surgery, Jiangmen Central Hospital, Jiangmen, 529000, Guangdong, China
| | - Yaoming He
- Department of Gastrointestinal Surgery, Jiangmen Central Hospital, Jiangmen, 529000, Guangdong, China
| | - Yu He
- National Drug Clinical Trial Institution, Jiangmen Central Hospital, Jiangmen, 529000, Guangdong, China
| | - Weijun Liang
- Department of Gastrointestinal Surgery, Jiangmen Central Hospital, Jiangmen, 529000, Guangdong, China
| | - Chaorong Zhou
- Department of Gastrointestinal Surgery, Jiangmen Central Hospital, Jiangmen, 529000, Guangdong, China
| | - Meimei Wu
- Clinical Experimental Center, Jiangmen Key Laboratory of Clinical Biobanks and Translational Research, Jiangmen Central Hospital, Jiangmen, 529000, Guangdong, China
| | - Zijie Meng
- Clinical Experimental Center, Jiangmen Key Laboratory of Clinical Biobanks and Translational Research, Jiangmen Central Hospital, Jiangmen, 529000, Guangdong, China
| | - Wanglin Li
- The First Affiliated Hospital, Jinan University, Guangzhou, 529000, Guangdong, China.
| | - Jie Cao
- The First Affiliated Hospital, Jinan University, Guangzhou, 529000, Guangdong, China.
| |
Collapse
|
10
|
Tian K, Ying Y, Huang J, Wu H, Wei C, Li L, Chen L, Wu L. The expression, immune infiltration, prognosis, and experimental validation of OSBPL family genes in liver cancer. BMC Cancer 2023; 23:244. [PMID: 36918840 PMCID: PMC10015719 DOI: 10.1186/s12885-023-10713-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Accepted: 03/07/2023] [Indexed: 03/16/2023] Open
Abstract
BACKGROUND Liver cancer is the third most deadly malignant tumor in the world with poor prognosis and lacks early diagnostic markers. It is urgent need to explore new biomarkers and prognostic factors. The oxysterol-binding protein-like family proteins (OSBPLs) are essential mediators of lipid transportation and cholesterol balancing which has been reported to participate in cancer progression. So far, the expression, immune infiltration, and prognosis of OSBPLs have not been elucidated in liver cancer. METHODS The differential expressions of OSBPLs between liver tumor and normal tissues were assessed by analyzing RNA-seq data from TCGA and protein data from CPTAC, respectively. Subsequently, genetic variations, potential functional enrichment analysis, and immune cell infiltration were analyzed. Further, the prognostic effects of OSBPLs were identified via constructing lasso models and performing receiver operating characteristic curve (ROC) analysis. Moreover, 10 local liver cancer specimens were involved to validate the expression of OSBPL3 via immunohistochemistry (IHC) assay. Finally, CCK-8, cell cycle, apoptosis, transwell assays, real time qPCR (RT-qPCR), and western blot assays were conducted to explore the function of OSBPL3 in liver cancer cells. RESULTS The mRNA of OSBPL2, OSBPL3, and OSBPL8 were highly expressed while OSBPL6 was lowly expressed in liver cancer samples compared with normal samples. As to the protein expression, OSBPL2 and OSBPL3 were significantly elevated and OSBPL5, OSBPL6, OSBPL9, OSBPL10, OSBPL11 were downregulated in tumor samples. A positive correlation was found between copy number variations (CNV) and the expression of OSBPL2, OSBPL8, OSBPL9, OSBPL11, while DNA methylation was negatively associated with the expressions of OSBPLs. Of these, CNV amplification mainly contributed to the overexpression of OSBPL2 and DNA methylation may be responsible for the high expression of OSBPL3. Interestingly, OSBPL3, OSBPL5, SOBPL7, and OSBPL10 were significantly positively correlated with immune infiltration. Notably, OSBPL3 was identified correlated to overall survival (OS) and disease specific survival (DSS) in liver cancer. Functionally, knocking down OSBPL3 reduced liver cancer cell viability, induced a G2/M cell cycle arrest, promoted apoptosis, and restrained cell migration. CONCLUSION In aggregate, we reported a heretofore undescribed role of OSBPLs in liver cancer by analyzing multi-omics data. Importantly, we identified OSBPL3 was overexpressed in liver tumor compared with normal and its high expression was correlated with poor OS and DSS. Inhibition of OSBPL3 resulted in a pronounced decrease in cell proliferation and migration.
Collapse
Affiliation(s)
- Kunpeng Tian
- School of Medicine, Guangxi University, Nanning, Guangxi, 530004, China
| | - Yongling Ying
- School of Medicine, Guangxi University, Nanning, Guangxi, 530004, China
| | - Jingjing Huang
- Department of Spleen and Stomach Liver Diseases, The First Affiliated Hospital of Guangxi University of Traditional Chinese Medicine, Nanning, Guangxi, 530200, China.,Guangxi Key Laboratory of Translational Medicine of Integrated Traditional Chinese and Western Medicine, Nanning, Guangxi, 530200, China.,Guangxi Key Laboratory of Molecular Biology of Preventive Medicine of Traditional Chinese Medicine, Nanning, Guangxi, 530200, China
| | - Hao Wu
- School of Medicine, Guangxi University, Nanning, Guangxi, 530004, China
| | - Chengyue Wei
- School of Medicine, Guangxi University, Nanning, Guangxi, 530004, China
| | - Liang Li
- School of Medicine, Guangxi University, Nanning, Guangxi, 530004, China
| | - Longjun Chen
- School of Medicine, Guangxi University, Nanning, Guangxi, 530004, China
| | - Lichuan Wu
- School of Medicine, Guangxi University, Nanning, Guangxi, 530004, China.
| |
Collapse
|
11
|
Kobayashi J, Arita M, Sakai S, Kojima H, Senda M, Senda T, Hanada K, Kato R. Ligand Recognition by the Lipid Transfer Domain of Human OSBP Is Important for Enterovirus Replication. ACS Infect Dis 2022; 8:1161-1170. [PMID: 35613096 DOI: 10.1021/acsinfecdis.2c00108] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Oxysterol-binding protein (OSBP), which transports cholesterol and phosphatidylinositol 4-monophosphate (PtdIns[4]P) between different organelles, serves as a conserved host factor for the replication of various viruses, and OSBP inhibitors exhibit antiviral effects. Here, we determined the crystal structure of the lipid transfer domain of human OSBP in complex with endogenous cholesterol. The hydrocarbon tail and tetracyclic ring of cholesterol interact with the hydrophobic tunnel of OSBP, and the hydroxyl group of cholesterol forms a hydrogen bond network at the bottom of the tunnel. Systematic mutagenesis of the ligand-binding region revealed that M446W and L590W substitutions confer functional tolerance to an OSBP inhibitor, T-00127-HEV2. Employing the M446W variant as a functional replacement for the endogenous OSBP in the presence of T-00127-HEV2, we have identified previously unappreciated amino acid residues required for viral replication. The combined use of the inhibitor and the OSBP variant will be useful in elucidating the enigmatic in vivo functions of OSBP.
Collapse
Affiliation(s)
- Jun Kobayashi
- Structural Biology Research Center, Institute of Materials Structure Science, High Energy Accelerator Research Organization (KEK), 1-1 Oho, Tsukuba, Ibaraki 305-0801, Japan
| | - Minetaro Arita
- Department of Virology II, National Institute of Infectious Diseases, 4-7-1 Gakuen, Musashimurayama, Tokyo 208-0011, Japan
| | - Shota Sakai
- Department of Biochemistry and Cell Biology, National Institute of Infectious Diseases, 1-23-1 Toyama, Shinjuku-ku, Tokyo 162-8640, Japan
| | - Hirotatsu Kojima
- Drug Discovery Initiative, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Miki Senda
- Structural Biology Research Center, Institute of Materials Structure Science, High Energy Accelerator Research Organization (KEK), 1-1 Oho, Tsukuba, Ibaraki 305-0801, Japan
| | - Toshiya Senda
- Structural Biology Research Center, Institute of Materials Structure Science, High Energy Accelerator Research Organization (KEK), 1-1 Oho, Tsukuba, Ibaraki 305-0801, Japan
| | - Kentaro Hanada
- Department of Biochemistry and Cell Biology, National Institute of Infectious Diseases, 1-23-1 Toyama, Shinjuku-ku, Tokyo 162-8640, Japan
| | - Ryuichi Kato
- Structural Biology Research Center, Institute of Materials Structure Science, High Energy Accelerator Research Organization (KEK), 1-1 Oho, Tsukuba, Ibaraki 305-0801, Japan
| |
Collapse
|
12
|
Quon E, Nenadic A, Zaman MF, Johansen J, Beh CT. ER-PM membrane contact site regulation by yeast ORPs and membrane stress pathways. PLoS Genet 2022; 18:e1010106. [PMID: 35239652 PMCID: PMC8923467 DOI: 10.1371/journal.pgen.1010106] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 03/15/2022] [Accepted: 02/16/2022] [Indexed: 02/01/2023] Open
Abstract
In yeast, at least seven proteins (Ice2p, Ist2p, Scs2/22p, Tcb1-Tcb3p) affect cortical endoplasmic reticulum (ER) tethering and contact with the plasma membrane (PM). In Δ-super-tether (Δ-s-tether) cells that lack these tethers, cortical ER-PM association is all but gone. Yeast OSBP homologue (Osh) proteins are also implicated in membrane contact site (MCS) assembly, perhaps as subunits for multicomponent tethers, though their function at MCSs involves intermembrane lipid transfer. Paradoxically, when analyzed by fluorescence and electron microscopy, the elimination of the OSH gene family does not reduce cortical ER-PM association but dramatically increases it. In response to the inactivation of all Osh proteins, the yeast E-Syt (extended-synaptotagmin) homologue Tcb3p is post-transcriptionally upregulated thereby generating additional Tcb3p-dependent ER-PM MCSs for recruiting more cortical ER to the PM. Although the elimination of OSH genes and the deletion of ER-PM tether genes have divergent effects on cortical ER-PM association, both elicit the Environmental Stress Response (ESR). Through comparisons of transcriptomic profiles of cells lacking OSH genes or ER-PM tethers, changes in ESR expression are partially manifested through the induction of the HOG (high-osmolarity glycerol) PM stress pathway or the ER-specific UPR (unfolded protein response) pathway, respectively. Defects in either UPR or HOG pathways also increase ER-PM MCSs, and expression of extra “artificial ER-PM membrane staples” rescues growth of UPR mutants challenged with lethal ER stress. Transcriptome analysis of OSH and Δ-s-tether mutants also revealed dysregulation of inositol-dependent phospholipid gene expression, and the combined lethality of osh4Δ and Δ-s-tether mutations is suppressed by overexpression of the phosphatidic acid biosynthetic gene, DGK1. These findings establish that the Tcb3p tether is induced by ER and PM stresses and ER-PM MCSs augment responses to membrane stresses, which are integrated through the broader ESR pathway. Membrane contact sites (MCSs) between the two largest cellular membranes, the endoplasmic reticulum (ER) and the plasma membrane (PM), are regulatory interfaces for lipid synthesis and bidirectional transport. The yeast Osh protein family, which represents the seven yeast oxysterol-binding protein related proteins (ORPs), is implicated in MCS regulation and lipid transfer between membranes. Ironically, we find that when all Osh proteins eliminated, ER-PM association is not reduced but significantly increases. We hypothesized this increase is due to compensatory increases in levels of tether proteins that physically link the ER and PM. In fact, in response to inactivating Osh protein expression, amounts of the tether protein Tcb3 increase and more ER-PM MCSs are produced. By testing the genomic transcriptional responses to the elimination of OSH and ER-PM tether genes, we find these mutants disrupt phospholipid regulation and they elicit the Environmental Stress Response (ESR) pathway, which integrates many different responses needed for recovery after cellular stress. OSH and ER-PM tether genes affect specific stress response pathways that impact the PM and ER, respectively. Combining OSH and tether mutations results in cell lethality, but these cells survive by increased expression of a key phospholipid biosynthetic gene. Based on these results, we propose that OSH and ER-PM tether genes affect phospholipid regulation and protect the PM and ER through membrane stress responses integrated through the ESR pathway.
Collapse
Affiliation(s)
- Evan Quon
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, British Columbia, Canada
| | - Aleksa Nenadic
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, British Columbia, Canada
| | - Mohammad F. Zaman
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, British Columbia, Canada
| | - Jesper Johansen
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, British Columbia, Canada
| | - Christopher T. Beh
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, British Columbia, Canada
- Centre for Cell Biology, Development, and Disease, Simon Fraser University, Burnaby, Canada
- * E-mail:
| |
Collapse
|
13
|
Potential Prognostic Biomarkers of OSBPL Family Genes in Patients with Pancreatic Ductal Adenocarcinoma. Biomedicines 2021; 9:biomedicines9111601. [PMID: 34829830 PMCID: PMC8615799 DOI: 10.3390/biomedicines9111601] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 10/24/2021] [Accepted: 10/26/2021] [Indexed: 12/24/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a highly fatal malignancy with poor survival outcomes. In addition, oxysterol-binding protein-like (OSBPL) family members are reported to be involved in lipid binding and transport and play critical roles in tumorigenesis. However, relationships between PDAC and OSBPL family members have not comprehensively been elucidated. In this study, we used the Oncomine and GEPIA 2 databases to analyze OSBPL transcription expressions in PDAC. The Kaplan–Meier plotter and TIMER 2.0 were used to assess the relationships between overall survival (OS) and immune-infiltration with OSBPL family members. Co-expression data from cBioPortal were downloaded to assess the correlated pathways with OSBPL gene family members using DAVID. The expressions of OSBPL3, OSBPL8, OSBPL10, and OSBPL11 were found to be highly upregulated in PDAC. Low expressions of OSBPL3, OSBPL8, and OSBPL10 indicated longer OS. The functions of OSBPL family members were mainly associated with several potential signaling pathways in cancer cells, including ATP binding, integrin binding, receptor binding, and the renin-angiotensin system (RAS) signaling pathway. The transcription levels of OSBPL gene family members were connected with several immune infiltrates. Collectively, OSBPL family members are influential biomarkers for the early diagnosis of PDAC and have prognostic value, with the promise of precise treatment of PDAC in the future.
Collapse
|
14
|
Reinmuth L, Hsiao CC, Hamann J, Rosenkilde M, Mackrill J. Multiple Targets for Oxysterols in Their Regulation of the Immune System. Cells 2021; 10:cells10082078. [PMID: 34440846 PMCID: PMC8391951 DOI: 10.3390/cells10082078] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 08/05/2021] [Accepted: 08/11/2021] [Indexed: 02/07/2023] Open
Abstract
Oxysterols, or cholesterol oxidation products, are naturally occurring lipids which regulate the physiology of cells, including those of the immune system. In contrast to effects that are mediated through nuclear receptors or by epigenetic mechanism, which take tens of minutes to occur, changes in the activities of cell-surface receptors caused by oxysterols can be extremely rapid, often taking place within subsecond timescales. Such cell-surface receptor effects of oxysterols allow for the regulation of fast cellular processes, such as motility, secretion and endocytosis. These cellular processes play critical roles in both the innate and adaptive immune systems. This review will survey the two broad classes of cell-surface receptors for oxysterols (G-protein coupled receptors (GPCRs) and ion channels), the mechanisms by which cholesterol oxidation products act on them, and their presence and functions in the different cell types of the immune system. Overall, this review will highlight the potential of oxysterols, synthetic derivatives and their receptors for physiological and therapeutic modulation of the immune system.
Collapse
Affiliation(s)
- Lisa Reinmuth
- Laboratory for Molecular Pharmacology, Department of Biomedical Sciences, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen, Denmark;
| | - Cheng-Chih Hsiao
- Department of Experimental Immunology, Amsterdam Institute for Infection and Immunity, Amsterdam University Medical Centers, Meibergdreef 9, 1105AZ Amsterdam, The Netherlands; (C.-C.H.); (J.H.)
- Neuroimmunology Research Group, The Netherlands Institute for Neuroscience, 1105BA Amsterdam, The Netherlands
| | - Jörg Hamann
- Department of Experimental Immunology, Amsterdam Institute for Infection and Immunity, Amsterdam University Medical Centers, Meibergdreef 9, 1105AZ Amsterdam, The Netherlands; (C.-C.H.); (J.H.)
- Neuroimmunology Research Group, The Netherlands Institute for Neuroscience, 1105BA Amsterdam, The Netherlands
| | - Mette Rosenkilde
- Laboratory for Molecular Pharmacology, Department of Biomedical Sciences, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen, Denmark;
- Correspondence: (M.R.); (J.M.); Tel.: +353-(0)21-490-1400 (J.M.)
| | - John Mackrill
- Department of Physiology, School of Medicine, BioSciences Institute, University College Cork, College Road, Cork T12 YT20, Ireland
- Correspondence: (M.R.); (J.M.); Tel.: +353-(0)21-490-1400 (J.M.)
| |
Collapse
|
15
|
Santos MF, Rappa G, Karbanová J, Fontana S, Bella MAD, Pope MR, Parrino B, Cascioferro SM, Vistoli G, Diana P, Cirrincione G, Arena GO, Woo G, Huang K, Huynh T, Moschetti M, Alessandro R, Corbeil D, Lorico A. Itraconazole inhibits nuclear delivery of extracellular vesicle cargo by disrupting the entry of late endosomes into the nucleoplasmic reticulum. J Extracell Vesicles 2021; 10:e12132. [PMID: 34429859 PMCID: PMC8363911 DOI: 10.1002/jev2.12132] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 07/21/2021] [Accepted: 07/26/2021] [Indexed: 01/01/2023] Open
Abstract
Extracellular vesicles (EVs) are mediators of intercellular communication under both healthy and pathological conditions, including the induction of pro-metastatic traits, but it is not yet known how and where functional cargoes of EVs are delivered to their targets in host cell compartments. We have described that after endocytosis, EVs reach Rab7+ late endosomes and a fraction of these enter the nucleoplasmic reticulum and transport EV biomaterials to the host cell nucleoplasm. Their entry therein and docking to outer nuclear membrane occur through a tripartite complex formed by the proteins VAP-A, ORP3 and Rab7 (VOR complex). Here, we report that the antifungal compound itraconazole (ICZ), but not its main metabolite hydroxy-ICZ or ketoconazole, disrupts the binding of Rab7 to ORP3-VAP-A complexes, leading to inhibition of EV-mediated pro-metastatic morphological changes including cell migration behaviour of colon cancer cells. With novel, smaller chemical drugs, inhibition of the VOR complex was maintained, although the ICZ moieties responsible for antifungal activity and interference with intracellular cholesterol distribution were removed. Knowing that cancer cells hijack their microenvironment and that EVs derived from them determine the pre-metastatic niche, small-sized inhibitors of nuclear transfer of EV cargo into host cells could find cancer therapeutic applications, particularly in combination with direct targeting of cancer cells.
Collapse
Affiliation(s)
- Mark F. Santos
- College of MedicineTouro University NevadaHendersonNevadaUSA
| | - Germana Rappa
- College of MedicineTouro University NevadaHendersonNevadaUSA
| | - Jana Karbanová
- Biotechnology Centre and Centre for Molecular and Cellular BioengineeringTechnische Universität DresdenDresdenGermany
| | - Simona Fontana
- Department of Biomedicine, Neurosciences and Advanced DiagnosticsUniversity of PalermoPalermoItaly
| | | | | | - Barbara Parrino
- Dipartimento di Scienze e Tecnologie BiologicheChimiche e FarmaceuticheUniversità degli Studi di PalermoPalermoItaly
| | - Stella Maria Cascioferro
- Dipartimento di Scienze e Tecnologie BiologicheChimiche e FarmaceuticheUniversità degli Studi di PalermoPalermoItaly
| | - Giulio Vistoli
- Dipartimento di Scienze FarmaceuticheUniversità degli Studi di MilanoMilanItaly
| | - Patrizia Diana
- Dipartimento di Scienze e Tecnologie BiologicheChimiche e FarmaceuticheUniversità degli Studi di PalermoPalermoItaly
| | - Girolamo Cirrincione
- Dipartimento di Scienze e Tecnologie BiologicheChimiche e FarmaceuticheUniversità degli Studi di PalermoPalermoItaly
| | - Goffredo O. Arena
- Department of SurgeryMcGill UniversityMontréalQuébecCanada
- Fondazione Istituto G. GiglioCefalùItaly
| | - Gyunghwi Woo
- College of MedicineTouro University NevadaHendersonNevadaUSA
| | - Kevin Huang
- College of MedicineTouro University NevadaHendersonNevadaUSA
| | - Tony Huynh
- College of MedicineTouro University NevadaHendersonNevadaUSA
| | - Marta Moschetti
- Department of Biomedicine, Neurosciences and Advanced DiagnosticsUniversity of PalermoPalermoItaly
| | - Riccardo Alessandro
- Department of Biomedicine, Neurosciences and Advanced DiagnosticsUniversity of PalermoPalermoItaly
- Institute for Biomedical Research and Innovation (IRIB)National Research Council (CNR)PalermoItaly
| | - Denis Corbeil
- Biotechnology Centre and Centre for Molecular and Cellular BioengineeringTechnische Universität DresdenDresdenGermany
| | - Aurelio Lorico
- College of MedicineTouro University NevadaHendersonNevadaUSA
- Mediterranean Institute of OncologyViagrandeItaly
| |
Collapse
|
16
|
Nakatsu F, Kawasaki A. Functions of Oxysterol-Binding Proteins at Membrane Contact Sites and Their Control by Phosphoinositide Metabolism. Front Cell Dev Biol 2021; 9:664788. [PMID: 34249917 PMCID: PMC8264513 DOI: 10.3389/fcell.2021.664788] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2021] [Accepted: 05/06/2021] [Indexed: 01/10/2023] Open
Abstract
Lipids must be correctly transported within the cell to the right place at the right time in order to be fully functional. Non-vesicular lipid transport is mediated by so-called lipid transfer proteins (LTPs), which contain a hydrophobic cavity that sequesters lipid molecules. Oxysterol-binding protein (OSBP)-related proteins (ORPs) are a family of LTPs known to harbor lipid ligands, such as cholesterol and phospholipids. ORPs act as a sensor or transporter of those lipid ligands at membrane contact sites (MCSs) where two different cellular membranes are closely apposed. In particular, a characteristic functional property of ORPs is their role as a lipid exchanger. ORPs mediate counter-directional transport of two different lipid ligands at MCSs. Several, but not all, ORPs transport their lipid ligand from the endoplasmic reticulum (ER) in exchange for phosphatidylinositol 4-phosphate (PI4P), the other ligand, on apposed membranes. This ORP-mediated lipid “countertransport” is driven by the concentration gradient of PI4P between membranes, which is generated by its kinases and phosphatases. In this review, we will discuss how ORP function is tightly coupled to metabolism of phosphoinositides such as PI4P. Recent progress on the role of ORP-mediated lipid transport/countertransport at multiple MCSs in cellular functions will be also discussed.
Collapse
Affiliation(s)
- Fubito Nakatsu
- Department of Neurochemistry and Molecular Cell Biology, Niigata University School of Medicine and Graduate School of Medical/Dental Sciences, Niigata, Japan
| | - Asami Kawasaki
- Department of Neurochemistry and Molecular Cell Biology, Niigata University School of Medicine and Graduate School of Medical/Dental Sciences, Niigata, Japan
| |
Collapse
|
17
|
Rathod J, Yen HC, Liang B, Tseng YY, Chen CS, Wu WS. YPIBP: A repository for phosphoinositide-binding proteins in yeast. Comput Struct Biotechnol J 2021; 19:3692-3707. [PMID: 34285772 PMCID: PMC8261538 DOI: 10.1016/j.csbj.2021.06.035] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Revised: 06/08/2021] [Accepted: 06/22/2021] [Indexed: 11/25/2022] Open
Abstract
Phosphoinositides (PIs) are a family of eight lipids consisting of phosphatidylinositol (PtdIns) and its seven phosphorylated forms. PIs have important regulatory functions in the cell including lipid signaling, protein transport, and membrane trafficking. Yeast has been recognized as a eukaryotic model system to study lipid-protein interactions. Hundreds of yeast PI-binding proteins have been identified, but this research knowledge remains scattered. Besides, the complete PI-binding spectrum and potential PI-binding domains have not been interlinked. No comprehensive databases are available to support the lipid-protein interaction research on phosphoinositides. Here we constructed the first knowledgebase of Yeast Phosphoinositide-Binding Proteins (YPIBP), a repository consisting of 679 PI-binding proteins collected from high-throughput proteome-array and lipid-array studies, QuickGO, and a rigorous literature mining. The YPIBP also contains protein domain information in categories of lipid-binding domains, lipid-related domains and other domains. The YPIBP provides search and browse modes along with two enrichment analyses (PI-binding enrichment analysis and domain enrichment analysis). An interactive visualization is given to summarize the PI-domain-protein interactome. Finally, three case studies were given to demonstrate the utility of YPIBP. The YPIBP knowledgebase consolidates the present knowledge and provides new insights of the PI-binding proteins by bringing comprehensive and in-depth interaction network of the PI-binding proteins. YPIBP is available at http://cosbi7.ee.ncku.edu.tw/YPIBP/.
Collapse
Key Words
- ANTH, AP180 N-terminal Homology
- BAR, Bin-Amphiphysin-Rvs
- CAFA, Critical Assessment of Functional Annotation
- CRAL-TRIO, cellular retinaldehyde-binding protein (CRALBP) and TRIO guanine exchange factor
- Cvt, Cytoplasm-to-vacuole targeting
- ENTH, Epsin N-terminal Homology
- FDR, False Discovery Rate
- FYVE, Fab 1 (yeast orthologue of PIKfyve), YOTB, Vac 1 (vesicle transport protein), and EEA1
- GO, Gene Ontology
- ITC, Isothermal Titration Calorimetry
- LBD, Lipid-Binding Domain
- LMPD, LIPID MAPS Proteome Database
- LMSD, LIPID MAPS Structure Database
- LRD, Lipid-Related Domain
- Lipid-binding domain
- OMIM, Online Mendelian Inheritance in Man
- OSBP, Oxysterol-Binding Protein
- PH, Pleckstrin Homology
- PI(3,4)P2, phosphatidylinositol-3,4-bisphosphate
- PI(3,4,5)P3, phosphatidylinositol-3,4,5-trisphosphate
- PI(3,5)P2, phosphatidylinositol-3,5-bisphosphate
- PI(4,5)P2, phosphatidylinositol-4,5-bisphosphate
- PI-binding protein
- PI3P, phosphatidylinositol-3-phosphate
- PI4P, phosphatidylinositol-4-phosphate
- PI5P, phosphatidylinositol-5-phosphate
- PIs, Phosphoinositides
- PMID, PubMed ID
- PX, Phox Homology
- Phosphatidylinositol (PtdIns)
- Phosphoinositides (PIs)
- PtdIns, Phosphatidylinositol
- QCM, Quartz Crystal Microbalance
- S. cerevisiae
- SNX, Sorting Nexin
- SPR, Surface Plasmon Resonance
- YPIBP, Yeast Phosphoinositide-Binding Proteins
- Yeast
Collapse
Affiliation(s)
- Jagat Rathod
- Department of Earth Sciences, College of Sciences, National Cheng Kung University, Tainan 701, Taiwan
| | - Han-Chen Yen
- Department of Electrical Engineering, College of Electrical Engineering and Computer Science, National Cheng Kung University, Tainan 701, Taiwan
| | - Biqing Liang
- Department of Earth Sciences, College of Sciences, National Cheng Kung University, Tainan 701, Taiwan
| | - Yan-Yuan Tseng
- Center for Molecular Medicine and Genetics, Wayne State University, School of Medicine, Detroit, MI 48201, USA
| | - Chien-Sheng Chen
- Department of Food Safety/Hygiene and Risk Management, College of Medicine, National Cheng Kung University, Tainan 701, Taiwan
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan 701, Taiwan
| | - Wei-Sheng Wu
- Department of Electrical Engineering, College of Electrical Engineering and Computer Science, National Cheng Kung University, Tainan 701, Taiwan
| |
Collapse
|
18
|
Luquain-Costaz C, Rabia M, Hullin-Matsuda F, Delton I. Bis(monoacylglycero)phosphate, an important actor in the host endocytic machinery hijacked by SARS-CoV-2 and related viruses. Biochimie 2020; 179:247-256. [PMID: 33159981 PMCID: PMC7642752 DOI: 10.1016/j.biochi.2020.10.018] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 10/28/2020] [Accepted: 10/29/2020] [Indexed: 12/12/2022]
Abstract
Viruses, including the novel coronavirus SARS-CoV-2, redirect infected cell metabolism to their own purposes. After binding to its receptor angiotensin-converting enzyme 2 (ACE2) on the cell surface, the SARS-CoV-2 is taken up by receptor-mediated endocytosis ending in the acidic endolysosomal compartment. The virus hijacks the endosomal machinery leading to fusion of viral and endosomal membranes and release of the viral RNA into the cytosol. This mini-review specifically highlights the membrane lipid organization of the endosomal system focusing on the unconventional and late endosome/lysosome-specific phospholipid, bis(monoacylglycero)phosphate (BMP). BMP is enriched in alveolar macrophages of lung, one of the target tissue of SARS-CoV-2. This review details the BMP structure, its unsaturated fatty acid composition and fusogenic properties that are essential for the highly dynamic formation of the intraluminal vesicles inside the endosomes. Interestingly, BMP is necessary for infection and replication of enveloped RNA virus such as SARS-CoV-1 and Dengue virus. We also emphasize the role of BMP in lipid sorting and degradation, especially cholesterol transport in cooperation with Niemann Pick type C proteins (NPC 1 and 2) and with some oxysterol-binding protein (OSBP)-related proteins (ORPs) as well as in sphingolipid degradation. Interestingly, numerous virus infection required NPC1 as well as ORPs along the endocytic pathway. Furthermore, BMP content is increased during pathological endosomal lipid accumulation in various lysosomal storage disorders. This is particularly important knowing the high percentage of patients with metabolic disorders among the SARS-CoV-2 infected patients presenting severe forms of COVID-19.
Collapse
Affiliation(s)
- Céline Luquain-Costaz
- Univ-Lyon, CarMeN Laboratory, Inserm U1060, INRAe U1397, INSA Lyon, Villeurbanne, France
| | - Maxence Rabia
- Univ-Lyon, CarMeN Laboratory, Inserm U1060, INRAe U1397, INSA Lyon, Villeurbanne, France
| | | | - Isabelle Delton
- Univ-Lyon, CarMeN Laboratory, Inserm U1060, INRAe U1397, INSA Lyon, Villeurbanne, France.
| |
Collapse
|
19
|
Zewe JP, Miller AM, Sangappa S, Wills RC, Goulden BD, Hammond GRV. Probing the subcellular distribution of phosphatidylinositol reveals a surprising lack at the plasma membrane. J Cell Biol 2020; 219:133808. [PMID: 32211893 PMCID: PMC7054989 DOI: 10.1083/jcb.201906127] [Citation(s) in RCA: 71] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Revised: 10/14/2019] [Accepted: 11/19/2019] [Indexed: 12/16/2022] Open
Abstract
The polyphosphoinositides (PPIn) are central regulatory lipids that direct membrane function in eukaryotic cells. Understanding how their synthesis is regulated is crucial to revealing these lipids’ role in health and disease. PPIn are derived from the major structural lipid, phosphatidylinositol (PI). However, although the distribution of most PPIn has been characterized, the subcellular localization of PI available for PPIn synthesis is not known. Here, we used several orthogonal approaches to map the subcellular distribution of PI, including localizing exogenous fluorescent PI, as well as detecting lipid conversion products of endogenous PI after acute chemogenetic activation of PI-specific phospholipase and 4-kinase. We report that PI is broadly distributed throughout intracellular membrane compartments. However, there is a surprising lack of PI in the plasma membrane compared with the PPIn. These experiments implicate regulation of PI supply to the plasma membrane, as opposed to regulation of PPIn-kinases, as crucial to the control of PPIn synthesis and function at the PM.
Collapse
Affiliation(s)
- James P Zewe
- Department of Cell Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - April M Miller
- Department of Cell Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Sahana Sangappa
- Department of Cell Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Rachel C Wills
- Department of Cell Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Brady D Goulden
- Department of Cell Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Gerald R V Hammond
- Department of Cell Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA
| |
Collapse
|
20
|
Corbeil D, Santos MF, Karbanová J, Kurth T, Rappa G, Lorico A. Uptake and Fate of Extracellular Membrane Vesicles: Nucleoplasmic Reticulum-Associated Late Endosomes as a New Gate to Intercellular Communication. Cells 2020; 9:cells9091931. [PMID: 32825578 PMCID: PMC7563309 DOI: 10.3390/cells9091931] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 08/18/2020] [Accepted: 08/20/2020] [Indexed: 02/06/2023] Open
Abstract
Extracellular membrane vesicles (EVs) are emerging as new vehicles in intercellular communication, but how the biological information contained in EVs is shared between cells remains elusive. Several mechanisms have been described to explain their release from donor cells and the initial step of their uptake by recipient cells, which triggers a cellular response. Yet, the intracellular routes and subcellular fate of EV content upon internalization remain poorly characterized. This is particularly true for EV-associated proteins and nucleic acids that shuttle to the nucleus of host cells. In this review, we will describe and discuss the release of EVs from donor cells, their uptake by recipient cells, and the fate of their cargoes, focusing on a novel intracellular route wherein small GTPase Rab7+ late endosomes containing endocytosed EVs enter into nuclear envelope invaginations and deliver their cargo components to the nucleoplasm of recipient cells. A tripartite protein complex composed of (VAMP)-associated protein A (VAP-A), oxysterol-binding protein (OSBP)-related protein-3 (ORP3), and Rab7 is essential for the transfer of EV-derived components to the nuclear compartment by orchestrating the particular localization of late endosomes in the nucleoplasmic reticulum.
Collapse
Affiliation(s)
- Denis Corbeil
- Biotechnology Center (BIOTEC) and Center for Molecular and Cellular Bioengineering (CMCB), Technische Universität Dresden, Tatzberg 47-49, 01307 Dresden, Germany; (J.K.)
- Correspondence: (D.C.); (A.L.); Tel.: +49-(0)351-463-40118 (D.C.); +1-(702)-777-3942 (A.L.); Fax: +49-(0)351-463-40244 (D.C.); +1-(702)-777-1758 (A.L.)
| | - Mark F. Santos
- College of Osteopathic Medicine, Touro University Nevada, 874 American Pacific Drive, Henderson, NV 89014, USA; (M.F.S.); (G.R.)
| | - Jana Karbanová
- Biotechnology Center (BIOTEC) and Center for Molecular and Cellular Bioengineering (CMCB), Technische Universität Dresden, Tatzberg 47-49, 01307 Dresden, Germany; (J.K.)
| | - Thomas Kurth
- Center for Regenerative Therapies Dresden and CMCB, Technische Universität Dresden, Fetscherstraße 105, 01307 Dresden, Germany; (T.K.)
| | - Germana Rappa
- College of Osteopathic Medicine, Touro University Nevada, 874 American Pacific Drive, Henderson, NV 89014, USA; (M.F.S.); (G.R.)
| | - Aurelio Lorico
- College of Osteopathic Medicine, Touro University Nevada, 874 American Pacific Drive, Henderson, NV 89014, USA; (M.F.S.); (G.R.)
- Mediterranean Institute of Oncology, Via Penninazzo, 11, 95029 Viagrande, Italy
- Correspondence: (D.C.); (A.L.); Tel.: +49-(0)351-463-40118 (D.C.); +1-(702)-777-3942 (A.L.); Fax: +49-(0)351-463-40244 (D.C.); +1-(702)-777-1758 (A.L.)
| |
Collapse
|
21
|
Zhang D. Interplay between endoplasmic reticulum membrane contacts and actomyosin cytoskeleton. Cytoskeleton (Hoboken) 2020; 77:241-248. [PMID: 32543125 DOI: 10.1002/cm.21623] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 06/11/2020] [Accepted: 06/11/2020] [Indexed: 12/13/2022]
Abstract
Eukaryotic membrane-bound organelles, exhibiting distinctive morphologies, dynamics and functions, are interconnected at membrane contact sites (MCSs) through numerous tethering machineries. MCSs are required for many fundamental cellular processes, such as non-vesicular lipid transfer, calcium transport and organelle homeostasis. Actin cytoskeleton and myosin motors are known to dynamically interact with different membrane boundaries, facilitating organelle movements and partitioning. Intriguingly, recent studies have pinpointed a special participation of actomyosin at various MCSs involving the endoplasmic reticulum (ER), the most extensive membranous organelle in the cell. Here, I summarize emerging roles of ER MCSs in modulating actomyosin structures and discuss feedback functions of such actomyosin regulation at these MCSs.
Collapse
Affiliation(s)
- Dan Zhang
- Temasek Life Sciences Laboratory, 1 Research Link, National University of Singapore, Singapore, Singapore.,Department of Biological Sciences, National University of Singapore, Singapore, Singapore
| |
Collapse
|
22
|
Analysis of Low Molecular Weight Substances and Related Processes Influencing Cellular Cholesterol Efflux. Pharmaceut Med 2020; 33:465-498. [PMID: 31933239 PMCID: PMC7101889 DOI: 10.1007/s40290-019-00308-w] [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] [Indexed: 02/08/2023]
Abstract
Cholesterol efflux is the key process protecting the vascular system from the development of atherosclerotic lesions. Various extracellular and intracellular events affect the ability of the cell to efflux excess cholesterol. To explore the possible pathways and processes that promote or inhibit cholesterol efflux, we applied a combined cheminformatic and bioinformatic approach. We performed a comprehensive analysis of published data on the various substances influencing cholesterol efflux and found 153 low molecular weight substances that are included in the Chemical Entities of Biological Interest (ChEBI) database. Pathway enrichment was performed for substances identified within the Reactome database, and 45 substances were selected in 93 significant pathways. The most common pathways included the energy-dependent processes related to active cholesterol transport from the cell, lipoprotein metabolism and lipid transport, and signaling pathways. The activators and inhibitors of cholesterol efflux were non-uniformly distributed among the different pathways: the substances influencing ‘biological oxidations’ activate cholesterol efflux and the substances influencing ‘Signaling by GPCR and PTK6’ inhibit efflux. This analysis may be used in the search and design of efflux effectors for therapies targeting structural and functional high-density lipoprotein deficiency.
Collapse
|
23
|
Pankratenko AV, Atabekova AK, Morozov SY, Solovyev AG. Membrane Contacts in Plasmodesmata: Structural Components and Their Functions. BIOCHEMISTRY (MOSCOW) 2020; 85:531-544. [DOI: 10.1134/s0006297920050028] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
24
|
D'Souza RS, Lim JY, Turgut A, Servage K, Zhang J, Orth K, Sosale NG, Lazzara MJ, Allegood J, Casanova JE. Calcium-stimulated disassembly of focal adhesions mediated by an ORP3/IQSec1 complex. eLife 2020; 9:54113. [PMID: 32234213 PMCID: PMC7159923 DOI: 10.7554/elife.54113] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Accepted: 03/30/2020] [Indexed: 12/15/2022] Open
Abstract
Coordinated assembly and disassembly of integrin-mediated focal adhesions (FAs) is essential for cell migration. Many studies have shown that FA disassembly requires Ca2+ influx, however our understanding of this process remains incomplete. Here, we show that Ca2+ influx via STIM1/Orai1 calcium channels, which cluster near FAs, leads to activation of the GTPase Arf5 via the Ca2+-activated GEF IQSec1, and that both IQSec1 and Arf5 activation are essential for adhesion disassembly. We further show that IQSec1 forms a complex with the lipid transfer protein ORP3, and that Ca2+ influx triggers PKC-dependent translocation of this complex to ER/plasma membrane (PM) contact sites adjacent to FAs. In addition to allosterically activating IQSec1, ORP3 also extracts PI4P from the PM, in exchange for phosphatidylcholine. ORP3-mediated lipid exchange is also important for FA turnover. Together, these findings identify a new pathway that links calcium influx to FA turnover during cell migration.
Collapse
Affiliation(s)
- Ryan S D'Souza
- Department of Cell Biology, University of Virginia Health System, Charlottesville, United States
| | - Jun Y Lim
- Department of Cell Biology, University of Virginia Health System, Charlottesville, United States
| | - Alper Turgut
- Department of Cell Biology, University of Virginia Health System, Charlottesville, United States
| | - Kelly Servage
- Department of Molecular Biology, University of Texas Southwest Medical Center, Dallas, United States.,Howard Hughes Medical Institute, Dallas, United States
| | - Junmei Zhang
- Department of Cell Biology, University of Virginia Health System, Charlottesville, United States
| | - Kim Orth
- Department of Molecular Biology, University of Texas Southwest Medical Center, Dallas, United States.,Howard Hughes Medical Institute, Dallas, United States
| | - Nisha G Sosale
- Department of Chemical Engineering, University of Virginia, Charlottesville, United States
| | - Matthew J Lazzara
- Department of Chemical Engineering, University of Virginia, Charlottesville, United States
| | - Jeremy Allegood
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University, Richmond, United States
| | - James E Casanova
- Department of Cell Biology, University of Virginia Health System, Charlottesville, United States
| |
Collapse
|
25
|
Sasaki K, Yoshida H. Golgi stress response and organelle zones. FEBS Lett 2019; 593:2330-2340. [PMID: 31344260 DOI: 10.1002/1873-3468.13554] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Revised: 07/21/2019] [Accepted: 07/22/2019] [Indexed: 12/29/2022]
Abstract
Organelles have been studied traditionally as single units, but a novel concept is now emerging: each organelle has distinct functional zones that regulate specific functions. The Golgi apparatus seems to have various zones, including zones for: glycosylphosphatidylinositol-anchored proteins; proteoglycan, mucin and lipid glycosylation; transport of cholesterol and ceramides; protein degradation (Golgi membrane-associated degradation); and signalling for apoptosis. The capacity for these specific functions and the size of the corresponding zones appear to be tightly regulated by the Golgi stress response to accommodate cellular demands. For instance, the proteoglycan and mucin zones seem to be separately augmented during the differentiation of chondrocytes and goblet cells, respectively. The mammalian Golgi stress response consists of several response pathways. The TFE3 pathway regulates the general function of the Golgi, such as structural maintenance, N-glycosylation and vesicular transport, whereas the proteoglycan pathway increases the expression of glycosylation enzymes for proteoglycans. The CREB3 and HSP47 pathways regulate pro- and anti-apoptotic functions, respectively. These observations indicate that the Golgi is a dynamic organelle, the capacity of which is upregulated according to cellular needs.
Collapse
Affiliation(s)
- Kanae Sasaki
- Department of Molecular Biochemistry, Graduate School of Life Science, University of Hyogo, Japan
| | - Hiderou Yoshida
- Department of Molecular Biochemistry, Graduate School of Life Science, University of Hyogo, Japan
| |
Collapse
|
26
|
Peng H, Yan Z, Zeng X, Zhang S, Jiang H, Huang H, Zhuo H. Serum and tissue proteomic signatures of patients with hepatocellular carcinoma using 2‑D gel electrophoresis. Mol Med Rep 2019; 20:1025-1038. [PMID: 31173207 PMCID: PMC6625405 DOI: 10.3892/mmr.2019.10311] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Accepted: 04/24/2019] [Indexed: 12/23/2022] Open
Abstract
Hepatocellular carcinoma (HCC) accounts for ~85% of primary liver cancer cases and is a leading cause of mortality worldwide. Effective early diagnosis is difficult for HCC; however, effective biomarkers may be beneficial for diagnosis. In the current study, serum samples, and HCC and adjacent tissue samples were obtained from patients with HCC for the detection of biomarkers using 2-D gel electrophoresis (2-DE) and matrix-assisted laser desorption/ionization-time of flight (TOF)/TOF mass spectrometry. The crude serum samples did not need to be prepared for removal of high abundance proteins. The mRNA expression levels of HCC-associated proteins were detected in tissues using reverse transcription-quantitative PCR. Statistical analysis and database matching were used to identify the differentially expressed proteins detected in the serum and tissue groups. Immunohistochemistry (IHC) was performed to detect the expression of significant proteins in HCC and adjacent tissues. The results revealed ~800 protein spots on a 2-DE gel that were detected in serum samples, and 1,200 spots were identified in the tissue samples. The protein and mRNA expression levels of oxysterol binding protein-like 11 (OSBPL11) in HCC serum and tissue samples were consistent. Pathway analysis demonstrated that members of the apolipoprotein family, particularly apolipoprotein E (APOE), and RAS family members were closely associated in HCC, either directly or via ferratin heavy polypeptide 1. IHC results demonstrated that the APOE protein serves an important role in liver cancer development. The lysis buffer used in the current study was effective for serum protein separation in 2-DE sample preparation. In addition, the present study revealed that downregulated OSBPL11 may be a potential indicator for HCC, and the apolipoprotein family, particularly APOE, and the RAS family may cooperatively serve an important role.
Collapse
Affiliation(s)
- Huifang Peng
- Department of Endocrinology, The First Affiliated Hospital and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, Henan 471000, P.R. China
| | - Zhijian Yan
- Department of Urology, The Affiliated Zhongshan Hospital, Xiamen University, Xiamen, Fujian 361004, P.R. China
| | - Xinhua Zeng
- State Key Laboratory of Stress Cell Biology, School of Life Science, Xiamen University, Xiamen, Fujian 361004, P.R. China
| | - Sheng Zhang
- Fujian Provincial Key Laboratory of Chronic Liver Disease and Hepatocellular Carcinoma, The Affiliated Zhongshan Hospital, Xiamen University, Xiamen, Fujian 361004, P.R. China
| | - Hongwei Jiang
- Department of Endocrinology, The First Affiliated Hospital and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, Henan 471000, P.R. China
| | - Heqing Huang
- State Key Laboratory of Stress Cell Biology, School of Life Science, Xiamen University, Xiamen, Fujian 361004, P.R. China
| | - Huiqin Zhuo
- Department of Gastrointestinal Surgery, Zhongshan Hospital, Xiamen University, Xiamen, Fujian 361004, P.R. China
| |
Collapse
|
27
|
Qiu S, Zeng B. Advances in understanding of the oxysterol-binding protein homologous in yeast and filamentous fungi. Int Microbiol 2019; 22:169-179. [PMID: 30810998 DOI: 10.1007/s10123-019-00056-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Revised: 01/02/2019] [Accepted: 01/03/2019] [Indexed: 01/14/2023]
Abstract
Oxysterol-binding protein is an important non-vesicular trafficking protein involved in the transportation of lipids in eukaryotic cells. Oxysterol-binding protein is identified as oxysterol-binding protein-related proteins (ORPs) in mammals and oxysterol-binding protein homologue (Osh) in yeast. Research has described the function and structure of oxysterol-binding protein in mammals and yeast, but little information about the protein's structure and function in filamentous fungi has been reported. This article focuses on recent advances in the research of Osh proteins in yeast and filamentous fungi, such as Aspergillus oryzae, Aspergillus nidulans, and Candida albicans. Furthermore, we point out some problems in the field, summarizing the membrane contact sites (MCS) of Osh proteins in yeast, and consider the future of Osh protein development.
Collapse
Affiliation(s)
- Shangkun Qiu
- Jiangxi Province Key Laboratory Bioprocess Engineering, Jiangxi Science and Technology Normal University, Nanchang, 330013, China
| | - Bin Zeng
- Jiangxi Province Key Laboratory Bioprocess Engineering, Jiangxi Science and Technology Normal University, Nanchang, 330013, China.
| |
Collapse
|
28
|
Petit JD, Immel F, Lins L, Bayer EM. Lipids or Proteins: Who Is Leading the Dance at Membrane Contact Sites? FRONTIERS IN PLANT SCIENCE 2019; 10:198. [PMID: 30846999 PMCID: PMC6393330 DOI: 10.3389/fpls.2019.00198] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Accepted: 02/05/2019] [Indexed: 05/19/2023]
Abstract
Understanding the mode of action of membrane contact sites (MCSs) across eukaryotic organisms at the near-atomic level to infer function at the cellular and tissue levels is a challenge scientists are currently facing. These peculiar systems dedicated to inter-organellar communication are perfect examples of cellular processes where the interplay between lipids and proteins is critical. In this mini review, we underline the link between membrane lipid environment, the recruitment of proteins at specialized membrane domains and the function of MCSs. More precisely, we want to give insights on the crucial role of lipids in defining the specificity of plant endoplasmic reticulum (ER)-plasma membrane (PM) MCSs and we further propose approaches to study them at multiple scales. Our goal is not so much to go into detailed description of MCSs, as there are numerous focused reviews on the subject, but rather try to pinpoint the critical elements defining those structures and give an original point of view by considering the subject from a near-atomic angle with a focus on lipids. We review current knowledge as to how lipids can define MCS territories, play a role in the recruitment and function of the MCS-associated proteins and in turn, how the lipid environment can be modified by proteins.
Collapse
Affiliation(s)
- Jules D. Petit
- UMR5200 CNRS, Laboratory of Membrane Biogenesis, University of Bordeaux, Villenave d’Ornon, France
- Laboratoire de Biophysique Moléculaire aux Interfaces, TERRA Research Centre, GX ABT, Université de Liège, Liège, Belgium
| | - Françoise Immel
- UMR5200 CNRS, Laboratory of Membrane Biogenesis, University of Bordeaux, Villenave d’Ornon, France
| | - Laurence Lins
- Laboratoire de Biophysique Moléculaire aux Interfaces, TERRA Research Centre, GX ABT, Université de Liège, Liège, Belgium
| | - Emmanuelle M. Bayer
- UMR5200 CNRS, Laboratory of Membrane Biogenesis, University of Bordeaux, Villenave d’Ornon, France
| |
Collapse
|
29
|
Tong J, Tan L, Chun C, Im YJ. Structural basis of human ORP1-Rab7 interaction for the late-endosome and lysosome targeting. PLoS One 2019; 14:e0211724. [PMID: 30721249 PMCID: PMC6363164 DOI: 10.1371/journal.pone.0211724] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Accepted: 01/20/2019] [Indexed: 02/07/2023] Open
Abstract
Oxysterol-binding protein (OSBP) and OSBP-related proteins (ORPs) constitute a family of lipid transfer proteins conserved in eukaryotes. ORP1 transports cholesterol at the interface between the late endosomes/lysosomes (LELs) and the endoplasmic reticulum (ER). ORP1 is targeted to the endosomal membranes by forming a tripartite complex with the LE GTPase Rab7 and its effector RILP (Rab7-interacting lysosomal protein). Here, we determined the crystal structure of human ORP1 ANK domain in complex with the GTP-bound form of Rab7. ORP1 ANK binds to the helix α3 of Rab7 located away from the switching regions, which makes the interaction independent of the nucleotide-binding state of Rab7. Thus, the effector-interacting switch regions of Rab7 are accessible for RILP binding, allowing formation of the ORP1-Rab7-RILP complex. ORP1 ANK binds to Rab7 and the Rab7-RILP complex with similar micro-molar affinities, which is consistent with the independence binding of ORP1 and RILP to Rab7. The structural model of the ORP1-Rab7-RILP complex correlates with the recruitment of ORP1 at the LEL-ER interface and the role in lipid transport and regulation.
Collapse
Affiliation(s)
- Junsen Tong
- College of Pharmacy, Chonnam National University, Gwangju, Republic of Korea
| | - Lingchen Tan
- College of Pharmacy, Chonnam National University, Gwangju, Republic of Korea
| | - ChangJu Chun
- College of Pharmacy, Chonnam National University, Gwangju, Republic of Korea
| | - Young Jun Im
- College of Pharmacy, Chonnam National University, Gwangju, Republic of Korea
- * E-mail:
| |
Collapse
|
30
|
Das K, Nozaki T. Non-vesicular Lipid Transport Machinery in Entamoeba histolytica. Front Cell Infect Microbiol 2018; 8:315. [PMID: 30283742 PMCID: PMC6156432 DOI: 10.3389/fcimb.2018.00315] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Accepted: 08/20/2018] [Indexed: 12/18/2022] Open
Abstract
Eukaryotic cells are organized into separate membrane-bound compartments that have specialized biochemical signature and function. Maintenance and regulation of distinct identity of each compartment is governed by the uneven distribution and intra-cellular movement of two essential biomolecules, lipids, and proteins. Non-vesicular lipid transport mediated by lipid transfer proteins plays a pivotal role in intra-cellular lipid trafficking and homeostasis whereas vesicular transport plays a central role in protein trafficking. Comparative study of lipid transport machinery in protist helps to better understand the pathogenesis and parasitism, and provides insight into eukaryotic evolution. Amebiasis, which is caused by Entamoeba histolytica, is one of the major enteric infections in humans, resulting in 40–100 thousand deaths annually. This protist has undergone remarkable alterations in the content and function of its sub-cellular compartments as well represented by its unique diversification of mitochondrion-related organelle, mitosome. We conducted domain-based search on AmoebaDB coupled with bioinformatics analyses and identified 22 potential lipid transfer protein homologs in E. histolytica, which are grouped into several sub-classes. Such in silico analyses have demonstrated the existence of well-organized lipid transport machinery in this parasite. We summarized and discussed the conservation and unique features of the whole repertoire of lipid transport proteins in E. histolytica.
Collapse
Affiliation(s)
- Koushik Das
- Graduate School of Medicine, The University of Tokyo, Bunkyō, Japan
| | - Tomoyoshi Nozaki
- Graduate School of Medicine, The University of Tokyo, Bunkyō, Japan
| |
Collapse
|
31
|
Intracellular and Plasma Membrane Events in Cholesterol Transport and Homeostasis. J Lipids 2018; 2018:3965054. [PMID: 30174957 PMCID: PMC6106919 DOI: 10.1155/2018/3965054] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Accepted: 07/26/2018] [Indexed: 12/13/2022] Open
Abstract
Cholesterol transport between intracellular compartments proceeds by both energy- and non-energy-dependent processes. Energy-dependent vesicular traffic partly contributes to cholesterol flux between endoplasmic reticulum, plasma membrane, and endocytic vesicles. Membrane contact sites and lipid transfer proteins are involved in nonvesicular lipid traffic. Only “active" cholesterol molecules outside of cholesterol-rich regions and partially exposed in water phase are able to fast transfer. The dissociation of partially exposed cholesterol molecules in water determines the rate of passive aqueous diffusion of cholesterol out of plasma membrane. ATP hydrolysis with concomitant conformational transition is required to cholesterol efflux by ABCA1 and ABCG1 transporters. Besides, scavenger receptor SR-B1 is involved also in cholesterol efflux by facilitated diffusion via hydrophobic tunnel within the molecule. Direct interaction of ABCA1 with apolipoprotein A-I (apoA-I) or apoA-I binding to high capacity binding sites in plasma membrane is important in cholesterol escape to free apoA-I. ABCG1-mediated efflux to fully lipidated apoA-I within high density lipoprotein particle proceeds more likely through the increase of “active” cholesterol level. Putative cholesterol-binding linear motifs within the structure of all three proteins ABCA1, ABCG1, and SR-B1 are suggested to contribute to the binding and transfer of cholesterol molecules from cytoplasmic to outer leaflets of lipid bilayer. Together, plasma membrane events and intracellular cholesterol metabolism and traffic determine the capacity of the cell for cholesterol efflux.
Collapse
|
32
|
Sobajima T, Yoshimura SI, Maeda T, Miyata H, Miyoshi E, Harada A. The Rab11-binding protein RELCH/KIAA1468 controls intracellular cholesterol distribution. J Cell Biol 2018; 217:1777-1796. [PMID: 29514919 PMCID: PMC5940305 DOI: 10.1083/jcb.201709123] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2017] [Revised: 01/20/2018] [Accepted: 02/16/2018] [Indexed: 12/29/2022] Open
Abstract
Cholesterol, which is endocytosed to the late endosome (LE)/lysosome, is delivered to other organelles through vesicular and nonvesicular transport mechanisms. In this study, we discuss a novel mechanism of cholesterol transport from recycling endosomes (REs) to the trans-Golgi network (TGN) through RELCH/KIAA1468, which is newly identified in this study as a Rab11-GTP- and OSBP-binding protein. After treating cells with 25-hydroxycholesterol to induce OSBP relocation from the cytoplasm to the TGN, REs accumulated around the TGN area, but this accumulation was diminished in RELCH- or OSBP-depleted cells. Cholesterol content in the TGN was decreased in Rab11-, RELCH-, and OSBP-depleted cells and increased in the LE/lysosome. According to in vitro reconstitution experiments, RELCH tethers Rab11-bound RE-like and OSBP-bound TGN-like liposomes and promotes OSBP-dependent cholesterol transfer from RE-like to TGN-like liposomes. These data suggest that RELCH promotes nonvesicular cholesterol transport from REs to the TGN through membrane tethering.
Collapse
Affiliation(s)
- Tomoaki Sobajima
- Department of Cell Biology, Graduate School of Medicine, Osaka University, Osaka, Japan
- Department of Molecular Biochemistry and Clinical Investigation, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Shin-Ichiro Yoshimura
- Department of Cell Biology, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Tomomi Maeda
- Department of Cell Biology, Graduate School of Medicine, Osaka University, Osaka, Japan
- Department of Molecular Biochemistry and Clinical Investigation, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Haruhiko Miyata
- Department of Experimental Genome Research, Research Institute for Microbial Diseases, Osaka, Japan
| | - Eiji Miyoshi
- Department of Molecular Biochemistry and Clinical Investigation, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Akihiro Harada
- Department of Cell Biology, Graduate School of Medicine, Osaka University, Osaka, Japan
| |
Collapse
|
33
|
Chang CL, Chen YJ, Liou J. ER-plasma membrane junctions: Why and how do we study them? BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2017; 1864:1494-1506. [PMID: 28554772 DOI: 10.1016/j.bbamcr.2017.05.018] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/09/2017] [Revised: 05/09/2017] [Accepted: 05/17/2017] [Indexed: 12/17/2022]
Abstract
Endoplasmic reticulum (ER)-plasma membrane (PM) junctions are membrane microdomains important for communication between the ER and the PM. ER-PM junctions were first reported in muscle cells in 1957, but mostly ignored in non-excitable cells due to their scarcity and lack of functional significance. In 2005, the discovery of stromal interaction molecule 1 (STIM1) mediating a universal Ca2+ feedback mechanism at ER-PM junctions in mammalian cells led to a resurgence of research interests toward ER-PM junctions. In the past decade, several major advancements have been made in this emerging topic in cell biology, including the generation of tools for labeling ER-PM junctions and the unraveling of mechanisms underlying regulation and functions of ER-PM junctions. This review summarizes early studies, recently developed tools, and current advances in the characterization and understanding of ER-PM junctions. This article is part of a Special Issue entitled: Membrane Contact Sites edited by Christian Ungermann and Benoit Kornmann.
Collapse
Affiliation(s)
- Chi-Lun Chang
- Department of Physiology, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Yu-Ju Chen
- Department of Physiology, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Jen Liou
- Department of Physiology, UT Southwestern Medical Center, Dallas, TX 75390, USA.
| |
Collapse
|
34
|
Wilhelm LP, Wendling C, Védie B, Kobayashi T, Chenard MP, Tomasetto C, Drin G, Alpy F. STARD3 mediates endoplasmic reticulum-to-endosome cholesterol transport at membrane contact sites. EMBO J 2017; 36:1412-1433. [PMID: 28377464 PMCID: PMC5430228 DOI: 10.15252/embj.201695917] [Citation(s) in RCA: 185] [Impact Index Per Article: 23.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2016] [Revised: 03/03/2017] [Accepted: 03/06/2017] [Indexed: 01/04/2023] Open
Abstract
StAR‐related lipid transfer domain‐3 (STARD3) is a sterol‐binding protein that creates endoplasmic reticulum (ER)–endosome contact sites. How this protein, at the crossroad between sterol uptake and synthesis pathways, impacts the intracellular distribution of this lipid was ill‐defined. Here, by using in situ cholesterol labeling and quantification, we demonstrated that STARD3 induces cholesterol accumulation in endosomes at the expense of the plasma membrane. STARD3‐mediated cholesterol routing depends both on its lipid transfer activity and its ability to create ER–endosome contacts. Corroborating this, in vitro reconstitution assays indicated that STARD3 and its ER‐anchored partner, Vesicle‐associated membrane protein‐associated protein (VAP), assemble into a machine that allows a highly efficient transport of cholesterol within membrane contacts. Thus, STARD3 is a cholesterol transporter scaffolding ER–endosome contacts and modulating cellular cholesterol repartition by delivering cholesterol to endosomes.
Collapse
Affiliation(s)
- Léa P Wilhelm
- Functional Genomics and Cancer Department, Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Illkirch, France.,Institut National de la Santé et de la Recherche Médicale (INSERM), U 964, Illkirch, France.,Centre National de la Recherche Scientifique (CNRS), UMR 7104, Illkirch, France.,Université de Strasbourg, Illkirch, France
| | - Corinne Wendling
- Functional Genomics and Cancer Department, Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Illkirch, France.,Institut National de la Santé et de la Recherche Médicale (INSERM), U 964, Illkirch, France.,Centre National de la Recherche Scientifique (CNRS), UMR 7104, Illkirch, France.,Université de Strasbourg, Illkirch, France
| | - Benoît Védie
- AP-HP (Assistance Publique - Hôpitaux de Paris), Hôpital Européen Georges Pompidou, Service de Biochimie, Paris, France
| | - Toshihide Kobayashi
- Université de Strasbourg, Illkirch, France.,Laboratory of Biophotonics and Pharmacology, Centre National de la Recherche Scientifique (CNRS), UMR 7213, Illkirch, France
| | - Marie-Pierre Chenard
- Functional Genomics and Cancer Department, Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Illkirch, France.,Université de Strasbourg, Illkirch, France.,Service d'Anatomie Pathologique Générale, Centre Hospitalier Universitaire de Hautepierre, Strasbourg, France
| | - Catherine Tomasetto
- Functional Genomics and Cancer Department, Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Illkirch, France .,Institut National de la Santé et de la Recherche Médicale (INSERM), U 964, Illkirch, France.,Centre National de la Recherche Scientifique (CNRS), UMR 7104, Illkirch, France.,Université de Strasbourg, Illkirch, France
| | - Guillaume Drin
- Université Côte d'Azur, CNRS Institut de Pharmacologie Moléculaire et Cellulaire, Valbonne, France
| | - Fabien Alpy
- Functional Genomics and Cancer Department, Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Illkirch, France .,Institut National de la Santé et de la Recherche Médicale (INSERM), U 964, Illkirch, France.,Centre National de la Recherche Scientifique (CNRS), UMR 7104, Illkirch, France.,Université de Strasbourg, Illkirch, France
| |
Collapse
|
35
|
Makowski SL, Tran TT, Field SJ. Emerging themes of regulation at the Golgi. Curr Opin Cell Biol 2017; 45:17-23. [PMID: 28213314 DOI: 10.1016/j.ceb.2017.01.004] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2016] [Accepted: 01/24/2017] [Indexed: 02/06/2023]
Abstract
The Golgi is generally recognized for its central role in the secretory pathway to orchestrate protein post-translational modification and trafficking of proteins and lipids to their final destination. Despite the common view of the Golgi as an inert sorting organelle, emerging data demonstrate that important signaling events occur at the Golgi, including those that regulate the trafficking function of the Golgi. The phosphatidylinositol-4-phosphate/GOLPH3/MYO18A/F-actin complex serves as a hub for signals that regulate Golgi trafficking function. Furthermore, the Golgi is increasingly appreciated for its important role in cell growth and in driving oncogenic transformation, as illuminated by the discovery that GOLPH3 and MYO18A are cancer drivers.
Collapse
Affiliation(s)
- Stefanie L Makowski
- Department of Medicine, Division of Endocrinology and Metabolism, University of California, San Diego, CA 92093-0707, USA
| | - Thuy Tt Tran
- Department of Medicine, Division of Endocrinology and Metabolism, University of California, San Diego, CA 92093-0707, USA
| | - Seth J Field
- Department of Medicine, Division of Endocrinology and Metabolism, University of California, San Diego, CA 92093-0707, USA.
| |
Collapse
|
36
|
Intracellular cholesterol transport proteins: roles in health and disease. Clin Sci (Lond) 2016; 130:1843-59. [DOI: 10.1042/cs20160339] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2016] [Accepted: 07/12/2016] [Indexed: 12/13/2022]
Abstract
Effective cholesterol homoeostasis is essential in maintaining cellular function, and this is achieved by a network of lipid-responsive nuclear transcription factors, and enzymes, receptors and transporters subject to post-transcriptional and post-translational regulation, whereas loss of these elegant, tightly regulated homoeostatic responses is integral to disease pathologies. Recent data suggest that sterol-binding sensors, exchangers and transporters contribute to regulation of cellular cholesterol homoeostasis and that genetic overexpression or deletion, or mutations, in a number of these proteins are linked with diseases, including atherosclerosis, dyslipidaemia, diabetes, congenital lipoid adrenal hyperplasia, cancer, autosomal dominant hearing loss and male infertility. This review focuses on current evidence exploring the function of members of the ‘START’ (steroidogenic acute regulatory protein-related lipid transfer) and ‘ORP’ (oxysterol-binding protein-related proteins) families of sterol-binding proteins in sterol homoeostasis in eukaryotic cells, and the evidence that they represent valid therapeutic targets to alleviate human disease.
Collapse
|
37
|
Park JS, Thorsness MK, Policastro R, McGoldrick LL, Hollingsworth NM, Thorsness PE, Neiman AM. Yeast Vps13 promotes mitochondrial function and is localized at membrane contact sites. Mol Biol Cell 2016; 27:2435-49. [PMID: 27280386 PMCID: PMC4966984 DOI: 10.1091/mbc.e16-02-0112] [Citation(s) in RCA: 129] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2016] [Accepted: 06/03/2016] [Indexed: 12/19/2022] Open
Abstract
Loss of VPS13 produces multiple phenotypes. This study implicates VPS13 in the function of membrane contact sites and suggests that different phenotypes of the mutant result from defects in different contact sites. In yeast, mutations found in the VPS13A gene of ChAc patients have specific defects in the mitochondrial aspect of VPS13 function. The Vps13 protein family is highly conserved in eukaryotic cells. Mutations in human VPS13 genes result in a variety of diseases, such as chorea acanthocytosis (ChAc), but the cellular functions of Vps13 proteins are not well defined. In yeast, there is a single VPS13 orthologue, which is required for at least two different processes: protein sorting to the vacuole and sporulation. This study demonstrates that VPS13 is also important for mitochondrial integrity. In addition to preventing transfer of DNA from the mitochondrion to the nucleus, VPS13 suppresses mitophagy and functions in parallel with the endoplasmic reticulum–mitochondrion encounter structure (ERMES). In different growth conditions, Vps13 localizes to endosome–mitochondrion contacts and to the nuclear–vacuole junctions, indicating that Vps13 may function at membrane contact sites. The ability of VPS13 to compensate for the absence of ERMES correlates with its intracellular distribution. We propose that Vps13 is present at multiple membrane contact sites and that separation-of-function mutants are due to loss of Vps13 at specific junctions. Introduction of VPS13A mutations identified in ChAc patients at cognate sites in yeast VPS13 are specifically defective in compensating for the lack of ERMES, suggesting that mitochondrial dysfunction might be the basis for ChAc.
Collapse
Affiliation(s)
- Jae-Sook Park
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, NY 11794-5215
| | - Mary K Thorsness
- Department of Molecular Biology, University of Wyoming, Laramie, WY 82071
| | - Robert Policastro
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, NY 11794-5215
| | - Luke L McGoldrick
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, NY 11794-5215
| | - Nancy M Hollingsworth
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, NY 11794-5215
| | - Peter E Thorsness
- Department of Molecular Biology, University of Wyoming, Laramie, WY 82071
| | - Aaron M Neiman
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, NY 11794-5215
| |
Collapse
|
38
|
Somerharju P. Is Spontaneous Translocation of Polar Lipids Between Cellular Organelles Negligible? Lipid Insights 2016; 8:87-93. [PMID: 27147824 PMCID: PMC4849424 DOI: 10.4137/lpi.s31616] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2016] [Revised: 02/21/2016] [Accepted: 02/22/2016] [Indexed: 01/23/2023] Open
Abstract
In most reviews addressing intracellular lipid trafficking, spontaneous diffusion of lipid monomers between the cellular organelles is considered biologically irrelevant because it is thought to be far too slow to significantly contribute to organelle biogenesis. This view is based on intervesicle transfer experiments carried out in vitro with few lipids as well as on the view that lipids are highly hydrophobic and thus cannot undergo spontaneous intermembrane diffusion at a significant rate. However, besides that single-chain lipids can translocate between vesicles in seconds, it has been demonstrated that the rate of spontaneous transfer of two-chain polar lipids can vary even 1000-fold, depending on the number of carbons and double bonds in the acyl chains. In addition, the rate of spontaneous lipid transfer can strongly depend on the experimental conditions such as vesicle composition and concentration. This review examines the studies suggesting that spontaneous lipid transfer is probably more relevant to intracellular trafficking of amphipathic lipids than commonly thought.
Collapse
Affiliation(s)
- Pentti Somerharju
- Department of Biochemistry and Developmental Biology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| |
Collapse
|
39
|
VAP, a Versatile Access Point for the Endoplasmic Reticulum: Review and analysis of FFAT-like motifs in the VAPome. Biochim Biophys Acta Mol Cell Biol Lipids 2016; 1861:952-961. [PMID: 26898182 DOI: 10.1016/j.bbalip.2016.02.009] [Citation(s) in RCA: 233] [Impact Index Per Article: 25.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2015] [Revised: 02/12/2016] [Accepted: 02/12/2016] [Indexed: 11/24/2022]
Abstract
Dysfunction of VAMP-associated protein (VAP) is associated with neurodegeneration, both Amyotrophic Lateral Sclerosis and Parkinson's disease. Here we summarize what is known about the intracellular interactions of VAP in humans and model organisms. VAP is a simple, small and highly conserved protein on the cytoplasmic face of the endoplasmic reticulum (ER). It is the sole protein on that large organelle that acts as a receptor for cytoplasmic proteins. This may explain the extremely wide range of interacting partners of VAP, with components of many cellular pathways binding it to access the ER. Many proteins that bind VAP also target other intracellular membranes, so VAP is a component of multiple molecular bridges at membrane contact sites between the ER and other organelles. So far approximately 100 proteins have been identified in the VAP interactome (VAPome), of which a small minority have a "two phenylalanines in an acidic tract" (FFAT) motif as it was originally defined. We have analyzed the entire VAPome in humans and yeast using a simple algorithm that identifies many more FFAT-like motifs. We show that approximately 50% of the VAPome binds directly or indirectly via the VAP-FFAT interaction. We also review evidence on pathogenesis in genetic disorders of VAP, which appear to arise from reduced overall VAP levels, leading to ER stress. It is not possible to identify one single interaction that underlies disease. This article is part of a Special Issue entitled: The cellular lipid landscape edited by Tim P. Levine and Anant K. Menon.
Collapse
|