1
|
Li M, Xing X, Yuan J, Zeng Z. Research progress on the regulatory role of cell membrane surface tension in cell behavior. Heliyon 2024; 10:e29923. [PMID: 38720730 PMCID: PMC11076917 DOI: 10.1016/j.heliyon.2024.e29923] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 04/17/2024] [Accepted: 04/17/2024] [Indexed: 05/12/2024] Open
Abstract
Cell membrane surface tension has emerged as a pivotal biophysical factor governing cell behavior and fate. This review systematically delineates recent advances in techniques for cell membrane surface tension quantification, mechanosensing mechanisms, and regulatory roles of cell membrane surface tension in modulating major cellular processes. Micropipette aspiration, tether pulling, and newly developed fluorescent probes enable the measurement of cell membrane surface tension with spatiotemporal precision. Cells perceive cell membrane surface tension via conduits including mechanosensitive ion channels, curvature-sensing proteins (e.g. BAR domain proteins), and cortex-membrane attachment proteins (e.g. ERM proteins). Through membrane receptors like integrins, cells convert mechanical cues into biochemical signals. This conversion triggers cytoskeletal remodeling and extracellular matrix interactions in response to environmental changes. Elevated cell membrane surface tension suppresses cell spreading, migration, and endocytosis while facilitating exocytosis. Moreover, reduced cell membrane surface tension promotes embryonic stem cell differentiation and cancer cell invasion, underscoring cell membrane surface tension as a regulator of cell plasticity. Outstanding questions remain regarding cell membrane surface tension regulatory mechanisms and roles in tissue development/disease in vivo. Emerging tools to manipulate cell membrane surface tension with high spatiotemporal control in combination with omics approaches will facilitate the elucidation of cell membrane surface tension-mediated effects on signaling networks across various cell types/states. This will accelerate the development of cell membrane surface tension-based biomarkers and therapeutics for regenerative medicine and cancer. Overall, this review provides critical insights into cell membrane surface tension as a potent orchestrator of cell function, with broader impacts across mechanobiology.
Collapse
Affiliation(s)
- Manqing Li
- School of Public Health, Sun Yat-sen University, Guangzhou, 5180080, China
| | - Xiumei Xing
- School of Public Health, Sun Yat-sen University, Guangzhou, 5180080, China
| | - Jianhui Yuan
- Nanshan District Center for Disease Control and Prevention, Shenzhen, 518054, China
| | - Zhuoying Zeng
- The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen University, Shenzhen, 518035, China
- Chemical Analysis & Physical Testing Institute, Shenzhen Center for Disease Control and Prevention, Shenzhen, 518055, China
| |
Collapse
|
2
|
van den Berg NWE, Kawasaki M, Nariswari FA, Fabrizi B, Neefs J, van der Made I, Wesselink R, van Boven WJP, Driessen AHG, Jongejan A, de Groot JR. MicroRNAs in atrial fibrillation target genes in structural remodelling. Cell Tissue Res 2023; 394:497-514. [PMID: 37833432 DOI: 10.1007/s00441-023-03823-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Accepted: 08/07/2023] [Indexed: 10/15/2023]
Abstract
We aim to elucidate how miRNAs regulate the mRNA signature of atrial fibrillation (AF), to gain mechanistic insight and identify candidate targets for future therapies. We present combined miRNA-mRNA sequencing using atrial tissues of patient without AF (n = 22), with paroxysmal AF (n = 22) and with persistent AF (n = 20). mRNA sequencing previously uncovered upregulated epithelial to mesenchymal transition, endothelial cell proliferation and extracellular matrix remodelling involving glycoproteins and proteoglycans in AF. MiRNA co-sequencing discovered miRNAs regulating the mRNA expression changes. Key downregulated miRNAs included miR-135b-5p, miR-138-5p, miR-200a-3p, miR-200b-3p and miR-31-5p and key upregulated miRNAs were miR-144-3p, miR-15b-3p, miR-182-5p miR-18b-5p, miR-4306 and miR-206. MiRNA expression levels were negatively correlated with the expression levels of a multitude of predicted target genes. Downregulated miRNAs associated with increased gene expression are involved in upregulated epithelial and endothelial cell migration and glycosaminoglycan biosynthesis. In vitro inhibition of miR-135b-5p and miR-138-5p validated an effect of miRNAs on multiple predicted targets. Altogether, the discovered miRNAs may be explored in further functional studies as potential targets for anti-fibrotic therapies in AF.
Collapse
Affiliation(s)
- Nicoline W E van den Berg
- Amsterdam UMC, University of Amsterdam, Heart Center; Department of Clinical and Experimental Cardiology and Cardiothoracic Surgery, Amsterdam Cardiovascular Sciences, Meibergdreef 9, 1105AZ, Amsterdam, The Netherlands.
| | - Makiri Kawasaki
- Amsterdam UMC, University of Amsterdam, Heart Center; Department of Clinical and Experimental Cardiology and Cardiothoracic Surgery, Amsterdam Cardiovascular Sciences, Meibergdreef 9, 1105AZ, Amsterdam, The Netherlands
| | - Fransisca A Nariswari
- Amsterdam UMC, University of Amsterdam, Heart Center; Department of Clinical and Experimental Cardiology and Cardiothoracic Surgery, Amsterdam Cardiovascular Sciences, Meibergdreef 9, 1105AZ, Amsterdam, The Netherlands
| | - Benedetta Fabrizi
- Amsterdam UMC, University of Amsterdam, Heart Center; Department of Clinical and Experimental Cardiology and Cardiothoracic Surgery, Amsterdam Cardiovascular Sciences, Meibergdreef 9, 1105AZ, Amsterdam, The Netherlands
| | - Jolien Neefs
- Amsterdam UMC, University of Amsterdam, Heart Center; Department of Clinical and Experimental Cardiology and Cardiothoracic Surgery, Amsterdam Cardiovascular Sciences, Meibergdreef 9, 1105AZ, Amsterdam, The Netherlands
| | - Ingeborg van der Made
- Amsterdam UMC, University of Amsterdam, Heart Center; Department of Clinical and Experimental Cardiology and Cardiothoracic Surgery, Amsterdam Cardiovascular Sciences, Meibergdreef 9, 1105AZ, Amsterdam, The Netherlands
| | - Robin Wesselink
- Amsterdam UMC, University of Amsterdam, Heart Center; Department of Clinical and Experimental Cardiology and Cardiothoracic Surgery, Amsterdam Cardiovascular Sciences, Meibergdreef 9, 1105AZ, Amsterdam, The Netherlands
| | - Wim Jan P van Boven
- Amsterdam UMC, University of Amsterdam, Heart Center; Department of Clinical and Experimental Cardiology and Cardiothoracic Surgery, Amsterdam Cardiovascular Sciences, Meibergdreef 9, 1105AZ, Amsterdam, The Netherlands
| | - Antoine H G Driessen
- Amsterdam UMC, University of Amsterdam, Heart Center; Department of Clinical and Experimental Cardiology and Cardiothoracic Surgery, Amsterdam Cardiovascular Sciences, Meibergdreef 9, 1105AZ, Amsterdam, The Netherlands
| | - Aldo Jongejan
- Amsterdam UMC, Department of Epidemiology and Data Science, University of Amsterdam, Meibergdreef 9, 1105AZ, Amsterdam, The Netherlands
| | - Joris R de Groot
- Amsterdam UMC, University of Amsterdam, Heart Center; Department of Clinical and Experimental Cardiology and Cardiothoracic Surgery, Amsterdam Cardiovascular Sciences, Meibergdreef 9, 1105AZ, Amsterdam, The Netherlands.
| |
Collapse
|
3
|
Franca R, Stocco G, Kiren V, Tessitore A, Fagioli F, Quarello P, Bertorello N, Rizzari C, Colombini A, Bettini LR, Locatelli F, Vinti L, Girardi K, Silvestri D, Valsecchi MG, Decorti G, Rabusin M. Impact of Mercaptopurine Metabolites on Disease Outcome in the AIEOP-BFM ALL 2009 Protocol for Acute Lymphoblastic Leukemia. Clin Pharmacol Ther 2023; 114:1082-1092. [PMID: 37550838 DOI: 10.1002/cpt.3022] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Accepted: 08/01/2023] [Indexed: 08/09/2023]
Abstract
In the maintenance phase of Associazione Italiana di Ematologia e Oncologia Pediatrica (AIEOP)- Berlin-Frankfurt-Muenster (BFM) acute lymphoblastic leukemia (ALL) 2009 protocol, mercaptopurine (MP) is given at the planned dose of 50 mg/m2 /day; however, dose adjustments are routinely performed to target patients' white blood cells to the optimal range of 2,000-3,000 cells/μL. Pediatric patients with ALL (n = 290, age: median (1st-3rd quartile): 4.8 (3.0-8.1) years; boys: 56.9%) were enrolled mainly in 4 medium-large Italian pediatric hospitals; 14.1% of patients relapsed after a median (1st-3rd quartile) follow-up time of 4.43 (3.82-5.46) years from maintenance beginning. MP metabolites (thionucleotide (TGN) and methyl-derivatives (MMPN)) were measured in the erythrocytes of 387 blood samples of 200 patients by high performance liquid chromatography with ultraviolet detection. Single-nucleotide polymorphisms (SNPs; (rs1800462, rs1800460, and rs1142345 in TPMT gene, rs116855232 in NUDT15, rs1127354, rs7270101, rs6051702 in ITPA, and rs2413739 in PACSIN2) were characterized by Taqman SNP genotyping assays. Cox proportional hazard models did not show an impact of TGN levels and variability on relapse. In contrast, after multivariate analysis, relapse hazard ratio (HR) increased in children with ALL of the intermediate risk arm compared with those in standard risk arm (3.44, 95% confidence interval (CI), 1.31-9.05, P = 0.012), and in carriers of the PACSIN2 rs2413739 T allele compared with those with the CC genotype (heterozygotes CT: HR, 2.32, 95% CI, 0.90-5.97, P = 0.081; and homozygous TT: HR, 4.14, 95% CI, 1.54-11.11, P = 0.005). Future studies are needed to confirm the lack of impact of TGN levels and variability on relapse in the AIEOP-BFM ALL trials, and to clarify the mechanism of PACSIN2 rs2413739 on outcome.
Collapse
Affiliation(s)
- Raffaella Franca
- Department of Medical, Surgical and Health Sciences, University of Trieste, Trieste, Italy
| | - Gabriele Stocco
- Department of Medical, Surgical and Health Sciences, University of Trieste, Trieste, Italy
- Institute for Maternal and Child Health - IRCCS Burlo Garofolo, Trieste, Italy
| | - Valentina Kiren
- Institute for Maternal and Child Health - IRCCS Burlo Garofolo, Trieste, Italy
| | - Antimo Tessitore
- Department of Medical, Surgical and Health Sciences, University of Trieste, Trieste, Italy
| | - Franca Fagioli
- Paediatric Onco-Haematology Department, Regina Margherita Children's Hospital, Turin, Italy
- Department of Public Health and Pediatrics, University of Turin, Turin, Italy
| | - Paola Quarello
- Paediatric Onco-Haematology Department, Regina Margherita Children's Hospital, Turin, Italy
- Department of Public Health and Pediatrics, University of Turin, Turin, Italy
| | - Nicoletta Bertorello
- Paediatric Onco-Haematology Department, Regina Margherita Children's Hospital, Turin, Italy
| | - Carmelo Rizzari
- University of Milano-Bicocca, Milan, Italy
- Pediatric Hematology Oncology Unit, MBBM Foundation, ASST Monza, Monza, Italy
| | - Antonella Colombini
- Pediatric Hematology Oncology Unit, MBBM Foundation, ASST Monza, Monza, Italy
| | - Laura Rachele Bettini
- University of Milano-Bicocca, Milan, Italy
- Pediatric Hematology Oncology Unit, MBBM Foundation, ASST Monza, Monza, Italy
| | - Franco Locatelli
- Pediatric Hematology and Oncology, IRCCS Ospedale Pediatrico Bambin Gesù, Rome, Italy
| | - Luciana Vinti
- Pediatric Hematology and Oncology, IRCCS Ospedale Pediatrico Bambin Gesù, Rome, Italy
| | - Katia Girardi
- Pediatric Hematology and Oncology, IRCCS Ospedale Pediatrico Bambin Gesù, Rome, Italy
| | - Daniela Silvestri
- Pediatric Hematology Oncology Unit, MBBM Foundation, ASST Monza, Monza, Italy
| | - Maria Grazia Valsecchi
- Bicocca Centre of Bioinformatics, Biostatistics and Bioimaging, School of Medicine and Surgery, University of Milano-Bicocca, Milan, Italy
| | - Giuliana Decorti
- Department of Medical, Surgical and Health Sciences, University of Trieste, Trieste, Italy
- Institute for Maternal and Child Health - IRCCS Burlo Garofolo, Trieste, Italy
| | - Marco Rabusin
- Institute for Maternal and Child Health - IRCCS Burlo Garofolo, Trieste, Italy
| |
Collapse
|
4
|
Bouslama R, Dumont V, Lindfors S, Paavolainen L, Tienari J, Nisen H, Mirtti T, Saleem MA, Gordin D, Groop PH, Suetsugu S, Lehtonen S. Phosphorylation of PACSIN2 at S313 Regulates Podocyte Architecture in Coordination with N-WASP. Cells 2023; 12:1487. [PMID: 37296607 PMCID: PMC10252800 DOI: 10.3390/cells12111487] [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: 01/06/2023] [Revised: 05/16/2023] [Accepted: 05/23/2023] [Indexed: 06/12/2023] Open
Abstract
Changes in the dynamic architecture of podocytes, the glomerular epithelial cells, lead to kidney dysfunction. Previous studies on protein kinase C and casein kinase 2 substrates in neurons 2 (PACSIN2), a known regulator of endocytosis and cytoskeletal organization, reveal a connection between PACSIN2 and kidney pathogenesis. Here, we show that the phosphorylation of PACSIN2 at serine 313 (S313) is increased in the glomeruli of rats with diabetic kidney disease. We found that phosphorylation at S313 is associated with kidney dysfunction and increased free fatty acids rather than with high glucose and diabetes alone. Phosphorylation of PACSIN2 emerged as a dynamic process that fine-tunes cell morphology and cytoskeletal arrangement, in cooperation with the regulator of the actin cytoskeleton, Neural Wiskott-Aldrich syndrome protein (N-WASP). PACSIN2 phosphorylation decreased N-WASP degradation while N-WASP inhibition triggered PACSIN2 phosphorylation at S313. Functionally, pS313-PACSIN2 regulated actin cytoskeleton rearrangement depending on the type of cell injury and the signaling pathways involved. Collectively, this study indicates that N-WASP induces phosphorylation of PACSIN2 at S313, which serves as a mechanism whereby cells regulate active actin-related processes. The dynamic phosphorylation of S313 is needed to regulate cytoskeletal reorganization.
Collapse
Affiliation(s)
- Rim Bouslama
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, 00290 Helsinki, Finland
| | - Vincent Dumont
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, 00290 Helsinki, Finland
| | - Sonja Lindfors
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, 00290 Helsinki, Finland
| | - Lassi Paavolainen
- Institute for Molecular Medicine Finland (FIMM), Helsinki Institute of Life Science (HiLIFE), University of Helsinki, 00290 Helsinki, Finland
| | - Jukka Tienari
- Department of Pathology, University of Helsinki, Helsinki, and Helsinki University Hospital, 05850 Hyvinkää, Finland
| | - Harry Nisen
- Department of Urology, Helsinki University Hospital, 00029 HUS, Finland
| | - Tuomas Mirtti
- Department of Pathology, Helsinki University Hospital, 00290 Helsinki, Finland
- Research Program in Systems Oncology, Faculty of Medicine, University of Helsinki, 00290 Helsinki, Finland
| | - Moin A. Saleem
- Children’s Renal Unit, Bristol Medical School, University of Bristol, Bristol BS8 1TS, UK
| | - Daniel Gordin
- Minerva Foundation Institute for Medical Research, 00290 Helsinki, Finland
- Abdominal Center, Nephrology, University of Helsinki and Helsinki University Hospital, 00290 Helsinki, Finland
- Joslin Diabetes Center, Harvard Medical School, Boston, MA 02215, USA
| | - Per-Henrik Groop
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, 00290 Helsinki, Finland
- Folkhälsan Institute of Genetics, Folkhälsan Research Center, 00290 Helsinki, Finland
- Department of Nephrology, University of Helsinki, Helsinki, and Helsinki University Hospital, 00290 Helsinki, Finland
- Department of Diabetes, Central Clinical School, Monash University, Melbourne, VIC 3800, Australia
| | - Shiro Suetsugu
- Division of Biological Science, Graduate School of Science and Technology, Nara Institute of Science and Technology, Ikoma 630-0192, Japan
- Data Science Center, Nara Institute of Science and Technology, Ikoma 630-0192, Japan
- Center for Digital Green-Innovation, Nara Institute of Science and Technology, Ikoma 630-0192, Japan
| | - Sanna Lehtonen
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, 00290 Helsinki, Finland
- Department of Pathology, University of Helsinki, 00290 Helsinki, Finland
| |
Collapse
|
5
|
Sotodosos-Alonso L, Pulgarín-Alfaro M, Del Pozo MA. Caveolae Mechanotransduction at the Interface between Cytoskeleton and Extracellular Matrix. Cells 2023; 12:cells12060942. [PMID: 36980283 PMCID: PMC10047380 DOI: 10.3390/cells12060942] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Revised: 03/08/2023] [Accepted: 03/10/2023] [Indexed: 03/30/2023] Open
Abstract
The plasma membrane (PM) is subjected to multiple mechanical forces, and it must adapt and respond to them. PM invaginations named caveolae, with a specific protein and lipid composition, play a crucial role in this mechanosensing and mechanotransduction process. They respond to PM tension changes by flattening, contributing to the buffering of high-range increases in mechanical tension, while novel structures termed dolines, sharing Caveolin1 as the main component, gradually respond to low and medium forces. Caveolae are associated with different types of cytoskeletal filaments, which regulate membrane tension and also initiate multiple mechanotransduction pathways. Caveolar components sense the mechanical properties of the substrate and orchestrate responses that modify the extracellular matrix (ECM) according to these stimuli. They perform this function through both physical remodeling of ECM, where the actin cytoskeleton is a central player, and via the chemical alteration of the ECM composition by exosome deposition. Here, we review mechanotransduction regulation mediated by caveolae and caveolar components, focusing on how mechanical cues are transmitted through the cellular cytoskeleton and how caveolae respond and remodel the ECM.
Collapse
Affiliation(s)
- Laura Sotodosos-Alonso
- Mechanoadaptation and Caveolae Biology Laboratory, Novel Mechanisms of Atherosclerosis Program, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), 28029 Madrid, Spain
| | - Marta Pulgarín-Alfaro
- Mechanoadaptation and Caveolae Biology Laboratory, Novel Mechanisms of Atherosclerosis Program, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), 28029 Madrid, Spain
| | - Miguel A Del Pozo
- Mechanoadaptation and Caveolae Biology Laboratory, Novel Mechanisms of Atherosclerosis Program, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), 28029 Madrid, Spain
| |
Collapse
|
6
|
Zudeh G, Franca R, Lucafò M, Bonten EJ, Bramuzzo M, Sgarra R, Lagatolla C, Franzin M, Evans WE, Decorti G, Stocco G. PACSIN2 as a modulator of autophagy and mercaptopurine cytotoxicity: mechanisms in lymphoid and intestinal cells. Life Sci Alliance 2023; 6:e202201610. [PMID: 36596605 PMCID: PMC9811133 DOI: 10.26508/lsa.202201610] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Revised: 12/14/2022] [Accepted: 12/15/2022] [Indexed: 01/05/2023] Open
Abstract
PACSIN2 variants are associated with gastrointestinal effects of thiopurines and thiopurine methyltransferase activity through an uncharacterized mechanism that is postulated to involve autophagy. This study aims to clarify the role of PACSIN2 in autophagy and in thiopurine cytotoxicity in leukemic and intestinal models. Higher autophagy and lower PACSIN2 levels were observed in inflamed compared with non-inflamed colon biopsies of inflammatory bowel disease pediatric patients at diagnosis. PACSIN2 was identified as an inhibitor of autophagy, putatively through inhibition of autophagosome formation by a protein-protein interaction with LC3-II, mediated by a LIR motif. Moreover, PACSIN2 resulted a modulator of mercaptopurine-induced cytotoxicity in intestinal cells, suggesting that PACSIN2-regulated autophagy levels might influence thiopurine sensitivity. However, PACSIN2 modulates cellular thiopurine methyltransferase activity via mechanisms distinct from its modulation of autophagy.
Collapse
Affiliation(s)
- Giulia Zudeh
- Department of Translational and Advanced Diagnostics, Institute for Maternal and Child Health I.R.C.C.S. Burlo Garofolo, Trieste, Italy
| | - Raffaella Franca
- Department of Medical, Surgical and Health Sciences, University of Trieste, Trieste, Italy
| | - Marianna Lucafò
- Department of Translational and Advanced Diagnostics, Institute for Maternal and Child Health I.R.C.C.S. Burlo Garofolo, Trieste, Italy
| | - Erik J Bonten
- Department of Chemical Biology and Therapeutics, Saint Jude Children's Research Hospital, Memphis, TN, USA
| | - Matteo Bramuzzo
- Department of Gastroenterology, Digestive Endoscopy and Nutrition Unit, Institute for Maternal and Child Health I.R.C.C.S. Burlo Garofolo, Trieste, Italy
| | - Riccardo Sgarra
- Department of Life Sciences, University of Trieste, Trieste, Italy
| | | | - Martina Franzin
- Department of Translational and Advanced Diagnostics, Institute for Maternal and Child Health I.R.C.C.S. Burlo Garofolo, Trieste, Italy
| | - William E Evans
- Department of Pharmaceutical Sciences, Saint Jude Children's Research Hospital, Memphis, TN, USA
| | - Giuliana Decorti
- Department of Translational and Advanced Diagnostics, Institute for Maternal and Child Health I.R.C.C.S. Burlo Garofolo, Trieste, Italy
- Department of Medical, Surgical and Health Sciences, University of Trieste, Trieste, Italy
| | - Gabriele Stocco
- Department of Translational and Advanced Diagnostics, Institute for Maternal and Child Health I.R.C.C.S. Burlo Garofolo, Trieste, Italy
- Department of Medical, Surgical and Health Sciences, University of Trieste, Trieste, Italy
| |
Collapse
|
7
|
Guo HL, Zhao YT, Wang WJ, Dong N, Hu YH, Zhang YY, Chen F, Zhou L, Li T. Optimizing thiopurine therapy in children with acute lymphoblastic leukemia: A promising “MINT” sequencing strategy and therapeutic “DNA-TG” monitoring. Front Pharmacol 2022; 13:941182. [PMID: 36238550 PMCID: PMC9552076 DOI: 10.3389/fphar.2022.941182] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Accepted: 08/02/2022] [Indexed: 11/13/2022] Open
Abstract
Thiopurines, including thioguanine (TG), 6-mercaptopurine (6-MP), and azathioprine (AZA), are extensively used in clinical practice in children with acute lymphoblastic leukemia (ALL) and inflammatory bowel diseases. However, the common adverse effects caused by myelosuppression and hepatotoxicity limit their application. Metabolizing enzymes such as thiopurine S-methyltransferase (TPMT), nudix hydrolase 15 (NUDT15), inosine triphosphate pyrophosphohydrolase (ITPA), and drug transporters like multidrug resistance-associated protein 4 (MRP4) have been reported to mediate the metabolism and transportation of thiopurine drugs. Hence, the single nucleotide polymorphisms (SNPs) in those genes could theoretically affect the pharmacokinetics and pharmacological effects of these drugs, and might also become one of the determinants of clinical efficacy and adverse effects. Moreover, long-term clinical practices have confirmed that thiopurine-related adverse reactions are associated with the systemic concentrations of their active metabolites. In this review, we mainly summarized the pharmacogenetic studies of thiopurine drugs. We also evaluated the therapeutic drug monitoring (TDM) research studies and focused on those active metabolites, hoping to continuously improve monitoring strategies for thiopurine therapy to maximize therapeutic efficacy and minimize the adverse effects or toxicity. We proposed that tailoring thiopurine dosing based on MRP4, ITPA, NUDT15, and TMPT genotypes, defined as “MINT” panel sequencing strategy, might contribute toward improving the efficacy and safety of thiopurines. Moreover, the DNA-incorporated thioguanine nucleotide (DNA-TG) metabolite level was more suitable for red cell 6-thioguanine nucleotide (6-TGNs) monitoring, which can better predict the efficacy and safety of thiopurines. Integrating the panel “MINT” sequencing strategy with therapeutic “DNA-TG” monitoring would offer a new insight into the precision thiopurine therapy for pediatric acute lymphoblastic leukemia patients.
Collapse
Affiliation(s)
- Hong-Li Guo
- Pharmaceutical Sciences Research Center, Department of Pharmacy, Children’s Hospital of Nanjing Medical University, Nanjing, China
| | - Yue-Tao Zhao
- School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
- Visiting Graduate Student from School of Basic Medicine and Clinical Pharmacy, Pharmaceutical University, Nanjing, China
| | - Wei-Jun Wang
- School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
- Visiting Graduate Student from School of Basic Medicine and Clinical Pharmacy, Pharmaceutical University, Nanjing, China
| | - Na Dong
- Institute of Pharmaceutical Science, China Pharmaceutical University, Nanjing, China
- School of Institute of Pharmaceutical Science, Pharmaceutical University, Nanjing, China
| | - Ya-Hui Hu
- Pharmaceutical Sciences Research Center, Department of Pharmacy, Children’s Hospital of Nanjing Medical University, Nanjing, China
| | - Yuan-Yuan Zhang
- Pharmaceutical Sciences Research Center, Department of Pharmacy, Children’s Hospital of Nanjing Medical University, Nanjing, China
| | - Feng Chen
- Pharmaceutical Sciences Research Center, Department of Pharmacy, Children’s Hospital of Nanjing Medical University, Nanjing, China
- *Correspondence: Feng Chen, ; Li Zhou, ; Tao Li,
| | - Li Zhou
- Hematology and Oncology Department, Children’s Hospital of Nanjing Medical University, Nanjing, China
- *Correspondence: Feng Chen, ; Li Zhou, ; Tao Li,
| | - Tao Li
- Department of Solid Oncology, Children’s Hospital of Nanjing Medical University, Nanjing, China
- *Correspondence: Feng Chen, ; Li Zhou, ; Tao Li,
| |
Collapse
|
8
|
Mallik B, Bhat S, Kumar V. Role of Bin‐Amphiphysin‐Rvs (BAR) domain proteins in mediating neuronal signaling and disease. Synapse 2022; 76:e22248. [DOI: 10.1002/syn.22248] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 05/13/2022] [Accepted: 07/18/2022] [Indexed: 11/06/2022]
Affiliation(s)
- Bhagaban Mallik
- Department of Biological Sciences Indian Institute of Science Education and Research (IISER) Bhopal Indore Bypass Road Bhopal Madhya Pradesh 462 066 India
| | - Sajad Bhat
- Department of Biological Sciences Indian Institute of Science Education and Research (IISER) Bhopal Indore Bypass Road Bhopal Madhya Pradesh 462 066 India
| | - Vimlesh Kumar
- Department of Biological Sciences Indian Institute of Science Education and Research (IISER) Bhopal Indore Bypass Road Bhopal Madhya Pradesh 462 066 India
| |
Collapse
|
9
|
Dumont V, Lehtonen S. PACSIN proteins in vivo: Roles in development and physiology. Acta Physiol (Oxf) 2022; 234:e13783. [PMID: 34990060 PMCID: PMC9285741 DOI: 10.1111/apha.13783] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 11/15/2021] [Accepted: 01/01/2022] [Indexed: 12/22/2022]
Abstract
Protein kinase C and casein kinase substrate in neurons (PACSINs), or syndapins (synaptic dynamin‐associated proteins), are a family of proteins involved in the regulation of cell cytoskeleton, intracellular trafficking and signalling. Over the last twenty years, PACSINs have been mostly studied in the in vitro and ex vivo settings, and only in the last decade reports on their function in vivo have emerged. We first summarize the identification, structure and cellular functions of PACSINs, and then focus on the relevance of PACSINs in vivo. During development in various model organisms, PACSINs participate in diverse processes, such as neural crest cell development, gastrulation, laterality development and neuromuscular junction formation. In mouse, PACSIN2 regulates angiogenesis during retinal development and in human, PACSIN2 associates with monosomy and embryonic implantation. In adulthood, PACSIN1 has been extensively studied in the brain and shown to regulate neuromorphogenesis, receptor trafficking and synaptic plasticity. Several genetic studies suggest a role for PACSIN1 in the development of schizophrenia, which is also supported by the phenotype of mice depleted of PACSIN1. PACSIN2 plays an essential role in the maintenance of intestinal homeostasis and participates in kidney repair processes after injury. PACSIN3 is abundant in muscle tissue and necessary for caveolar biogenesis to create membrane reservoirs, thus controlling muscle function, and has been linked to certain genetic muscular disorders. The above examples illustrate the importance of PACSINs in diverse physiological or tissue repair processes in various organs, and associations to diseases when their functions are disturbed.
Collapse
Affiliation(s)
- Vincent Dumont
- Department of Pathology and Research Program for Clinical and Molecular Metabolism Faculty of Medicine University of Helsinki Helsinki Finland
| | - Sanna Lehtonen
- Department of Pathology and Research Program for Clinical and Molecular Metabolism Faculty of Medicine University of Helsinki Helsinki Finland
- Department of Pathology University of Helsinki Helsinki Finland
| |
Collapse
|
10
|
Deo N, Redpath G. Serotonin Receptor and Transporter Endocytosis Is an Important Factor in the Cellular Basis of Depression and Anxiety. Front Cell Neurosci 2022; 15:804592. [PMID: 35280519 PMCID: PMC8912961 DOI: 10.3389/fncel.2021.804592] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Accepted: 12/16/2021] [Indexed: 11/13/2022] Open
Abstract
Depression and anxiety are common, debilitating psychiatric conditions affecting millions of people throughout the world. Current treatments revolve around selective serotonin reuptake inhibitors (SSRIs), yet these drugs are only moderately effective at relieving depression. Moreover, up to 30% of sufferers are SSRI non-responders. Endocytosis, the process by which plasma membrane and extracellular constituents are internalized into the cell, plays a central role in the regulation of serotonin (5-hydroxytryptophan, 5-HT) signaling, SSRI function and depression and anxiety pathogenesis. Despite their therapeutic potential, surprisingly little is known about the endocytosis of the serotonin receptors (5-HT receptors) or the serotonin transporter (SERT). A subset of 5-HT receptors are endocytosed by clathrin-mediated endocytosis following serotonin binding, while for the majority of 5-HT receptors the endocytic regulation is not known. SERT internalizes serotonin from the extracellular space into the cell to limit the availability of serotonin for receptor binding and signaling. Endocytosis of SERT reduces serotonin uptake, facilitating serotonin signaling. SSRIs predominantly inhibit SERT, preventing serotonin uptake to enhance 5-HT receptor signaling, while hallucinogenic compounds directly activate specific 5-HT receptors, altering their interaction with endocytic adaptor proteins to induce alternate signaling outcomes. Further, multiple polymorphisms and transcriptional/proteomic alterations have been linked to depression, anxiety, and SSRI non-response. In this review, we detail the endocytic regulation of 5-HT receptors and SERT and outline how SSRIs and hallucinogenic compounds modulate serotonin signaling through endocytosis. Finally, we will examine the deregulated proteomes in depression and anxiety and link these with 5-HT receptor and SERT endocytosis. Ultimately, in attempting to integrate the current studies on the cellular biology of depression and anxiety, we propose that endocytosis is an important factor in the cellular basis of depression and anxiety. We will highlight how a thorough understanding 5-HT receptor and SERT endocytosis is integral to understanding the biological basis of depression and anxiety, and to facilitate the development of a next generation of specific, efficacious antidepressant treatments.
Collapse
Affiliation(s)
- Nikita Deo
- Department of Biochemistry, University of Otago, Dunedin, New Zealand
| | - Gregory Redpath
- European Molecular Biology Lab (EMBL) Australia Node in Single Molecule Science, School of Medical Sciences and the Australian Research Council (ARC) Centre of Excellence in Advanced Molecular Imaging, University of New South Wales, Sydney, NSW, Australia
- *Correspondence: Gregory Redpath
| |
Collapse
|
11
|
Electrostatic Forces Mediate the Specificity of RHO GTPase-GDI Interactions. Int J Mol Sci 2021; 22:ijms222212493. [PMID: 34830380 PMCID: PMC8622166 DOI: 10.3390/ijms222212493] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Revised: 11/15/2021] [Accepted: 11/16/2021] [Indexed: 01/13/2023] Open
Abstract
Three decades of research have documented the spatiotemporal dynamics of RHO family GTPase membrane extraction regulated by guanine nucleotide dissociation inhibitors (GDIs), but the interplay of the kinetic mechanism and structural specificity of these interactions is as yet unresolved. To address this, we reconstituted the GDI-controlled spatial segregation of geranylgeranylated RHO protein RAC1 in vitro. Various biochemical and biophysical measurements provided unprecedented mechanistic details for GDI function with respect to RHO protein dynamics. We determined that membrane extraction of RHO GTPases by GDI occurs via a 3-step mechanism: (1) GDI non-specifically associates with the switch regions of the RHO GTPases; (2) an electrostatic switch determines the interaction specificity between the C-terminal polybasic region of RHO GTPases and two distinct negatively-charged clusters of GDI1; (3) a non-specific displacement of geranylgeranyl moiety from the membrane sequesters it into a hydrophobic cleft, effectively shielding it from the aqueous milieu. This study substantially extends the model for the mechanism of GDI-regulated RHO GTPase extraction from the membrane, and could have implications for clinical studies and drug development.
Collapse
|
12
|
Zudeh G, Franca R, Stocco G, Decorti G. Biomarkers for gastrointestinal adverse events related to thiopurine therapy. World J Gastroenterol 2021; 27:6348-6356. [PMID: 34720526 PMCID: PMC8517779 DOI: 10.3748/wjg.v27.i38.6348] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Revised: 04/29/2021] [Accepted: 09/03/2021] [Indexed: 02/06/2023] Open
Abstract
Thiopurines are immunomodulators used in the treatment of acute lymphoblastic leukemia and inflammatory bowel diseases. Adverse reactions to these agents are one of the main causes of treatment discontinuation or interruption. Myelosuppression is the most frequent adverse effect; however, approximately 5%-20% of patients develop gastrointestinal toxicity. The identification of biomarkers able to prevent and/or monitor these adverse reactions would be useful for clinicians for the proactive management of long-term thiopurine therapy. In this editorial, we discuss evidence supporting the use of PACSIN2, RAC1, and ITPA genes, in addition to TPMT and NUDT15, as possible biomarkers for thiopurine-related gastrointestinal toxicity.
Collapse
Affiliation(s)
- Giulia Zudeh
- Department of Life Sciences, University of Trieste, Trieste 34127, Italy
| | - Raffaella Franca
- Department of Medical, Surgical and Health Sciences, University of Trieste, Trieste 34149, Italy
| | - Gabriele Stocco
- Department of Life Sciences, University of Trieste, Trieste 34127, Italy
| | - Giuliana Decorti
- Department of Medical, Surgical and Health Sciences, University of Trieste, Trieste 34149, Italy
- Institute for Maternal and Child Health I.R.C.C.S Burlo Garofolo, Trieste 34137, Italy
| |
Collapse
|
13
|
Intrinsically disordered proteins and membranes: a marriage of convenience for cell signalling? Biochem Soc Trans 2021; 48:2669-2689. [PMID: 33155649 PMCID: PMC7752083 DOI: 10.1042/bst20200467] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 10/05/2020] [Accepted: 10/08/2020] [Indexed: 02/07/2023]
Abstract
The structure-function paradigm has guided investigations into the molecules involved in cellular signalling for decades. The peripheries of this paradigm, however, start to unravel when considering the co-operation between proteins and the membrane in signalling processes. Intrinsically disordered regions hold distinct advantages over folded domains in terms of their binding promiscuity, sensitivity to their particular environment and their ease of modulation through post-translational modifications. Low sequence complexity and bias towards charged residues are also favourable for the multivalent electrostatic interactions that occur at the surfaces of lipid bilayers. This review looks at the principles behind the successful marriage between protein disorder and membranes in addition to the role of this partnership in modifying and regulating signalling in cellular processes. The HVR (hypervariable region) of small GTPases is highlighted as a well-studied example of the nuanced role a short intrinsically disordered region can play in the fine-tuning of signalling pathways.
Collapse
|
14
|
Recent developments in membrane curvature sensing and induction by proteins. Biochim Biophys Acta Gen Subj 2021; 1865:129971. [PMID: 34333084 DOI: 10.1016/j.bbagen.2021.129971] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Revised: 07/11/2021] [Accepted: 07/25/2021] [Indexed: 12/22/2022]
Abstract
BACKGROUND Membrane-bound intracellular organelles have characteristic shapes attributed to different local membrane curvatures, and these attributes are conserved across species. Over the past decade, it has been confirmed that specific proteins control the large curvatures of the membrane, whereas many others due to their specific structural features can sense the curvatures and bind to the specific geometrical cues. Elucidating the interplay between sensing and induction is indispensable to understand the mechanisms behind various biological processes such as vesicular trafficking and budding. SCOPE OF REVIEW We provide an overview of major classes of membrane proteins and the mechanisms of curvature sensing and induction. We then discuss the importance of membrane elastic characteristics to induce the membrane shapes similar to intracellular organelles. Finally, we survey recently available assays developed for studying the curvature sensing and induction by many proteins. MAJOR CONCLUSIONS Recent theoretical/computational modeling along with experimental studies have uncovered fascinating connections between lipid membrane and protein interactions. However, the phenomena of protein localization and synchronization to generate spatiotemporal dynamics in membrane morphology are yet to be fully understood. GENERAL SIGNIFICANCE The understanding of protein-membrane interactions is essential to shed light on various biological processes. This further enables the technological applications of many natural proteins/peptides in therapeutic treatments. The studies of membrane dynamic shapes help to understand the fundamental functions of membranes, while the medicinal roles of various macromolecules (such as proteins, peptides, etc.) are being increasingly investigated.
Collapse
|
15
|
Dynamin 2 and BAR domain protein pacsin 2 cooperatively regulate formation and maturation of podosomes. Biochem Biophys Res Commun 2021; 571:145-151. [PMID: 34325130 DOI: 10.1016/j.bbrc.2021.07.041] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Accepted: 07/11/2021] [Indexed: 11/20/2022]
Abstract
Podosomes are actin-rich adhesion structures formed in a variety of cell types, such as monocytic cells or cancer cells, to facilitate attachment to and degradation of the extracellular matrix (ECM). Previous studies showed that dynamin 2, a large GTPase involved in membrane remodeling and actin organization, is required for podosome function. However, precise roles of dynamin 2 at the podosomes remain to be elucidated. In this study, we identified a BAR (Bin-Amphiphysin-Rvs167) domain protein pacsin 2 as a functional partner of dynamin 2 at podosomes. Dynamin 2 and pacsin 2 interact and co-localize to podosomes in Src-transformed NIH 3T3 (NIH-Src) cells. RNAi of either dynamin 2 or pacsin 2 in NIH-Src cells inhibited podosome formation and maturation, suggesting essential and related roles at podosomes. Consistently, RNAi of pacsin 2 prevented dynamin 2 localization to podosomes, and reciprocal RNAi of dynamin 2 prevented pacsin 2 localization to podosomes. Taking these results together, we conclude that dynamin 2 and pacsin 2 co-operatively regulate organization of podosomes in NIH-Src cells.
Collapse
|
16
|
Application of a C. trachomatis expression system to identify C. pneumoniae proteins translocated into host cells. J Bacteriol 2021; 203:JB.00511-20. [PMID: 33685970 PMCID: PMC8117517 DOI: 10.1128/jb.00511-20] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Chlamydia pneumoniae is a Gram-negative, obligate intracellular pathogen that causes community-acquired respiratory infections. C. pneumoniae uses a cell contact-dependent type-III secretion (T3S) system to translocate pathogen effector proteins that manipulate host cellular functions. While several C. pneumoniae T3S effectors have been proposed, few have been experimentally confirmed in Chlamydia In this study, we expressed 382 C. pneumoniae genes in C. trachomatis as fusion proteins to an epitope tag derived from glycogen synthase kinase 3β (GSK3β) which is the target of phosphorylation by mammalian kinases. Based on the detection of the tagged C. pneumoniae protein with anti-phospho GSK3β antibodies, we identified 49 novel C. pneumoniae-specific proteins that are translocated by C. trachomatis into the host cytoplasm and thus likely play a role as modifiers of host cellular functions. In this manner, we identified and characterized a new C. pneumoniae effector CPj0678 that recruits the host cell protein PACSIN2 to the plasma membrane. These findings indicate that C. trachomatis provides a powerful screening system to detect candidate effector proteins encoded by other pathogenic and endosymbiotic Chlamydia species.Importance Chlamydia injects numerous effector proteins into host cells to manipulate cellular functions important for bacterial survival. Based on findings in C. trachomatis, it has been proposed that between 5-10% of the C. pneumoniae genome, a related respiratory pathogen, encodes secreted effectors. However only a few C. pneumoniae effectors have been identified and experimentally validated. With the development of methods for the stable genetic transformation of C. trachomatis, it is now possible to use C. trachomatis shuttle plasmids to express and explore the function of proteins from other Chlamydia in a more relevant bacterial system. In this study, we experimentally determined that 49 C. pneumoniae-specific proteins are translocated into the host cytoplasm by Chlamydia secretion systems, and identify a novel effector required to recruit the host factor PACSIN2 to the plasma membrane during infection.
Collapse
|
17
|
Vreeken D, Bruikman CS, Stam W, Cox SML, Nagy Z, Zhang H, Postma RJ, van Zonneveld AJ, Hovingh GK, van Gils JM. Downregulation of Endothelial Plexin A4 Under Inflammatory Conditions Impairs Vascular Integrity. Front Cardiovasc Med 2021; 8:633609. [PMID: 34017863 PMCID: PMC8129156 DOI: 10.3389/fcvm.2021.633609] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Accepted: 03/31/2021] [Indexed: 12/30/2022] Open
Abstract
Objective: Besides hyperlipidemia, inflammation is an important determinant in the initiation and the progression of atherosclerosis. As Neuroimmune Guidance Cues (NGCs) are emerging as regulators of atherosclerosis, we set out to investigate the expression and function of inflammation-regulated NGCs. Methods and results: NGC expression in human monocytes and endothelial cells was assessed using a publicly available RNA dataset. Next, the mRNA levels of expressed NGCs were analyzed in primary human monocytes and endothelial cells after stimulation with IL1β or TNFα. Upon stimulation a total of 14 and 19 NGCs in monocytes and endothelial cells, respectively, were differentially expressed. Since plexin A4 (PLXNA4) was strongly downregulated in endothelial cells under inflammatory conditions, the role of PLXNA4 in endothelial function was investigated. Knockdown of PLXNA4 in endothelial cells markedly impaired the integrity of the monolayer leading to more elongated cells with an inflammatory phenotype. In addition, these cells showed an increase in actin stress fibers and decreased cell-cell junctions. Functional assays revealed decreased barrier function and capillary network formation of the endothelial cells, while vascular leakage and trans-endothelial migration of monocytes was increased. Conclusion: The current study demonstrates that pro-inflammatory conditions result in differential expression of NGCs in endothelial cells and monocytes, both culprit cell types in atherosclerosis. Specifically, endothelial PLXNA4 is reduced upon inflammation, while PLXNA4 maintains endothelial barrier function thereby preventing vascular leakage of fluids as well as cells. Taken together, PLXNA4 may well have a causal role in atherogenesis that deserves further investigation.
Collapse
Affiliation(s)
- Dianne Vreeken
- Department of Internal Medicine (Nephrology) and the Einthoven Laboratory for Vascular and Regenerative Medicine, Leiden University Medical Center, Leiden, Netherlands
| | - Caroline Suzanne Bruikman
- Amsterdam Cardiovascular Sciences, Department of Vascular Medicine, Amsterdam UMC, Amsterdam, Netherlands
| | - Wendy Stam
- Department of Internal Medicine (Nephrology) and the Einthoven Laboratory for Vascular and Regenerative Medicine, Leiden University Medical Center, Leiden, Netherlands
| | - Stefan Martinus Leonardus Cox
- Department of Internal Medicine (Nephrology) and the Einthoven Laboratory for Vascular and Regenerative Medicine, Leiden University Medical Center, Leiden, Netherlands
| | - Zsófia Nagy
- Department of Internal Medicine (Nephrology) and the Einthoven Laboratory for Vascular and Regenerative Medicine, Leiden University Medical Center, Leiden, Netherlands
| | - Huayu Zhang
- Department of Internal Medicine (Nephrology) and the Einthoven Laboratory for Vascular and Regenerative Medicine, Leiden University Medical Center, Leiden, Netherlands
| | - Rudmer Johannes Postma
- Department of Internal Medicine (Nephrology) and the Einthoven Laboratory for Vascular and Regenerative Medicine, Leiden University Medical Center, Leiden, Netherlands
| | - Anton Jan van Zonneveld
- Department of Internal Medicine (Nephrology) and the Einthoven Laboratory for Vascular and Regenerative Medicine, Leiden University Medical Center, Leiden, Netherlands
| | - Gerard Kornelis Hovingh
- Amsterdam Cardiovascular Sciences, Department of Vascular Medicine, Amsterdam UMC, Amsterdam, Netherlands.,Novo Nordisk A/S, Copenhagen, Denmark
| | - Janine Maria van Gils
- Department of Internal Medicine (Nephrology) and the Einthoven Laboratory for Vascular and Regenerative Medicine, Leiden University Medical Center, Leiden, Netherlands
| |
Collapse
|
18
|
Kapoor T, Dubey P, Shirolikar S, Ray K. An actomyosin clamp assembled by the Amphiphysin-Rho1-Dia/DAAM-Rok pathway reinforces somatic cell membrane folded around spermatid heads. Cell Rep 2021; 34:108918. [PMID: 33789114 DOI: 10.1016/j.celrep.2021.108918] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Revised: 12/22/2020] [Accepted: 03/08/2021] [Indexed: 12/14/2022] Open
Abstract
Membrane curvature recruits Bin-Amphiphysin-Rvs (BAR)-domain proteins and induces local F-actin assembly, which further modifies the membrane curvature and dynamics. The downstream molecular pathway in vivo is still unclear. Here, we show that a tubular endomembrane scaffold supported by contractile actomyosin stabilizes the somatic cyst cell membrane folded around rigid spermatid heads during the final stages of sperm maturation in Drosophila testis. The structure resembles an actin "basket" covering the bundle of spermatid heads. Genetic analyses suggest that the actomyosin organization is nucleated exclusively by the formins - Diaphanous and Dishevelled Associated Activator of Morphogenesis (DAAM) - downstream of Rho1, which is recruited by the BAR-domain protein Amphiphysin. Actomyosin activity at the actin basket gathers the spermatid heads into a compact bundle and resists the somatic cell invasion by intruding spermatids. These observations reveal a distinct response mechanism of actin-membrane interactions, which generates a cell-adhesion-like strategy through active clamping.
Collapse
Affiliation(s)
- Tushna Kapoor
- Department of Biological Sciences, Tata Institute of Fundamental Research, Mumbai 400005, India
| | - Pankaj Dubey
- Department of Biological Sciences, Tata Institute of Fundamental Research, Mumbai 400005, India
| | - Seema Shirolikar
- Department of Biological Sciences, Tata Institute of Fundamental Research, Mumbai 400005, India
| | - Krishanu Ray
- Department of Biological Sciences, Tata Institute of Fundamental Research, Mumbai 400005, India.
| |
Collapse
|
19
|
Chen K, Jiao X, Ashton A, Di Rocco A, Pestell TG, Sun Y, Zhao J, Casimiro MC, Li Z, Lisanti MP, McCue PA, Shen D, Achilefu S, Rui H, Pestell RG. The membrane-associated form of cyclin D1 enhances cellular invasion. Oncogenesis 2020; 9:83. [PMID: 32948740 PMCID: PMC7501870 DOI: 10.1038/s41389-020-00266-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Revised: 08/22/2020] [Accepted: 09/02/2020] [Indexed: 02/07/2023] Open
Abstract
The essential G1-cyclin, CCND1, is a collaborative nuclear oncogene that is frequently overexpressed in cancer. D-type cyclins bind and activate CDK4 and CDK6 thereby contributing to G1–S cell-cycle progression. In addition to the nucleus, herein cyclin D1 was also located in the cytoplasmic membrane. In contrast with the nuclear-localized form of cyclin D1 (cyclin D1NL), the cytoplasmic membrane-localized form of cyclin D1 (cyclin D1MEM) induced transwell migration and the velocity of cellular migration. The cyclin D1MEM was sufficient to induce G1–S cell-cycle progression, cellular proliferation, and colony formation. The cyclin D1MEM was sufficient to induce phosphorylation of the serine threonine kinase Akt (Ser473) and augmented extranuclear localized 17β-estradiol dendrimer conjugate (EDC)-mediated phosphorylation of Akt (Ser473). These studies suggest distinct subcellular compartments of cell cycle proteins may convey distinct functions.
Collapse
Affiliation(s)
- Ke Chen
- Department of Cancer Biology, Thomas Jefferson University, Philadelphia, PA, 19107, USA
| | - Xuanmao Jiao
- Pennsylvania Cancer and Regenerative Medicine Research Center, Baruch S. Blumberg Institute, Pennsylvania Biotechnology Center, Wynnewood, PA, 19096, USA
| | - Anthony Ashton
- Pennsylvania Cancer and Regenerative Medicine Research Center, Baruch S. Blumberg Institute, Pennsylvania Biotechnology Center, Wynnewood, PA, 19096, USA
| | - Agnese Di Rocco
- Pennsylvania Cancer and Regenerative Medicine Research Center, Baruch S. Blumberg Institute, Pennsylvania Biotechnology Center, Wynnewood, PA, 19096, USA
| | - Timothy G Pestell
- Department of Cancer Biology, Thomas Jefferson University, Philadelphia, PA, 19107, USA
| | - Yunguang Sun
- Department of Pathology, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
| | - Jun Zhao
- Pennsylvania Cancer and Regenerative Medicine Research Center, Baruch S. Blumberg Institute, Pennsylvania Biotechnology Center, Wynnewood, PA, 19096, USA
| | - Mathew C Casimiro
- Pennsylvania Cancer and Regenerative Medicine Research Center, Baruch S. Blumberg Institute, Pennsylvania Biotechnology Center, Wynnewood, PA, 19096, USA.,Dept of Science and Math, Abraham Baldwin Agricultural college, Tifton, GA, 31794, Georgia
| | - Zhiping Li
- Pennsylvania Cancer and Regenerative Medicine Research Center, Baruch S. Blumberg Institute, Pennsylvania Biotechnology Center, Wynnewood, PA, 19096, USA
| | - Michael P Lisanti
- Biomedical Research Centre (BRC), Translational Medicine, School of Environment and Life Sciences, University of Salford, Manchester, United Kingdom
| | - Peter A McCue
- Department of Pathology, Anatomy and Cell Biology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, 19107, USA
| | - Duanwen Shen
- Departments of Biomedical Engineering, Washington University, St. Louis, MO, 63110, USA
| | - Samuel Achilefu
- Departments of Biomedical Engineering, Washington University, St. Louis, MO, 63110, USA.,Departments of Radiology, Washington University, St. Louis, MO, 63110, USA.,Departments of Biochemistry & Molecular Biophysics, Washington University, St. Louis, MO, 63110, USA
| | - Hallgeir Rui
- Department of Pathology, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
| | - Richard G Pestell
- Pennsylvania Cancer and Regenerative Medicine Research Center, Baruch S. Blumberg Institute, Pennsylvania Biotechnology Center, Wynnewood, PA, 19096, USA. .,The Wistar Cancer Center, Wistar Institute, Philadelphia, PA, 19104, USA.
| |
Collapse
|
20
|
Tagging and Deleting of Endogenous Caveolar Components Using CRISPR/Cas9 Technology. Methods Mol Biol 2020. [PMID: 32548827 DOI: 10.1007/978-1-0716-0732-9_14] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register]
Abstract
Here, we describe how to utilize CRISPR/Cas9 technology in the generation of tissue culture cells with fluorescently tagged caveolar components as well as cells deleted of endogenous caveolar components. As one example, we will describe tagging of EHD2, caveolar neck protein, with Green Fluorescent protein (eGFP) from endogenous loci (knock-in, KI). As another example, we will describe deletion (knock-out, KO) of Caveolin1 (Cav1), an essential caveolar component in NIH/3T3 cells. In both instances, the modifications were achieved by using Cas9 delivery on plasmid DNA by electroporation and by utilizing FACS cell sorting for selection or enrichment of edited population of cells. We also provide a list with tested gRNA sequences to successfully produce KI and KO of other caveolar components.
Collapse
|
21
|
Nguyen LP, Tran SC, Suetsugu S, Lim YS, Hwang SB. PACSIN2 Interacts with Nonstructural Protein 5A and Regulates Hepatitis C Virus Assembly. J Virol 2020; 94:e01531-19. [PMID: 31801866 PMCID: PMC7022371 DOI: 10.1128/jvi.01531-19] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Accepted: 11/26/2019] [Indexed: 12/12/2022] Open
Abstract
Hepatitis C virus (HCV) is a major etiologic agent of chronic liver diseases. HCV is highly dependent on cellular machinery for viral propagation. Using protein microarray analysis, we previously identified 90 cellular proteins as nonstructural 5A (NS5A) interacting partners. Of these, protein kinase C and casein kinase substrate in neurons protein 2 (PACSIN2) was selected for further study. PACSIN2 belongs to the PACSIN family, which is involved in the formation of caveolae. Protein interaction between NS5A and PACSIN2 was confirmed by pulldown assay and further verified by both coimmunoprecipitation and immunofluorescence assays. We showed that PACSIN2 interacted with domain I of NS5A and the Fer-CIP4 homology (FCH)-Bin/amphiphysin/Rvs (F-BAR) region of PACSIN2. Interestingly, NS5A specifically attenuated protein kinase C alpha (PKCα)-mediated phosphorylation of PACSIN2 at serine 313 by interrupting PACSIN2 and PKCα interaction. In fact, mutation of the serine 313 to alanine (S313A) of PACSIN2 increased protein interaction with NS5A. Silencing of PACSIN2 decreased both viral RNA and protein expression levels of HCV. Ectopic expression of the small interfering RNA (siRNA)-resistant PACSIN2 recovered the viral infectivity, suggesting that PACSIN2 was specifically required for HCV propagation. PACSIN2 was involved in viral assembly without affecting other steps of the HCV life cycle. Indeed, overexpression of PACSIN2 promoted NS5A and core protein (core) interaction. We further showed that inhibition of PKCα increased NS5A and core interaction, suggesting that phosphorylation of PACSIN2 might influence HCV assembly. Moreover, PACSIN2 was required for lipid droplet formation via modulating extracellular signal-regulated kinase 1/2 (ERK1/2) phosphorylation. Taken together, these data indicate that HCV modulates PACSIN2 via NS5A to promote virion assembly.IMPORTANCE PACSIN2 is a lipid-binding protein that triggers the tubulation of the phosphatidic acid-containing membranes. The functional involvement of PACSIN2 in the virus life cycle has not yet been demonstrated. We showed that phosphorylation of PACSIN2 displayed a negative effect on NS5A and core interaction. The most significant finding is that NS5A prevents PKCα from binding to PACSIN2. Therefore, the phosphorylation level of PACSIN2 is decreased in HCV-infected cells. We showed that HCV NS5A interrupted PKCα-mediated PACSIN2 phosphorylation at serine 313, thereby promoting NS5A-PACSIN2 interaction. We further demonstrated that PACSIN2 modulated lipid droplet formation through ERK1/2 phosphorylation. These data provide evidence that PACSIN2 is a proviral cellular factor required for viral propagation.
Collapse
Affiliation(s)
- Lap P Nguyen
- Laboratory of RNA Viral Diseases, Korea Zoonosis Research Institute, Chonbuk National University, Iksan, South Korea
- National Research Laboratory of Hepatitis C Virus and Ilsong Institute of Life Science, Hallym University, Anyang, South Korea
| | - Si C Tran
- National Research Laboratory of Hepatitis C Virus and Ilsong Institute of Life Science, Hallym University, Anyang, South Korea
| | - Shiro Suetsugu
- Laboratory of Molecular Medicine and Cell Biology, Graduate School of Biosciences, Nara Institute of Science and Technology, Ikoma, Japan
| | - Yun-Sook Lim
- Laboratory of RNA Viral Diseases, Korea Zoonosis Research Institute, Chonbuk National University, Iksan, South Korea
- National Research Laboratory of Hepatitis C Virus and Ilsong Institute of Life Science, Hallym University, Anyang, South Korea
| | - Soon B Hwang
- Laboratory of RNA Viral Diseases, Korea Zoonosis Research Institute, Chonbuk National University, Iksan, South Korea
- National Research Laboratory of Hepatitis C Virus and Ilsong Institute of Life Science, Hallym University, Anyang, South Korea
| |
Collapse
|
22
|
PACSIN2 rs2413739 influence on thiopurine pharmacokinetics: validation studies in pediatric patients. THE PHARMACOGENOMICS JOURNAL 2019; 20:415-425. [PMID: 31792371 DOI: 10.1038/s41397-019-0130-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Revised: 11/09/2019] [Accepted: 11/20/2019] [Indexed: 12/11/2022]
Abstract
The aim of the study was to validate the impact of the single-nucleotide polymorphism rs2413739 (T > C) in the PACSIN2 gene on thiopurines pharmacological parameters and clinical response in an Italian cohort of pediatric patients with acute lymphoblastic leukemia (ALL) and inflammatory bowel disease (IBD). In ALL, PACSIN2 rs2413739 T allele was associated with a significant reduction of TPMT activity in erythrocytes (p = 0.0094, linear mixed-effect model, multivariate analysis considering TPMT genotype) and increased severe gastrointestinal toxicity during consolidation therapy (p = 0.049). A similar trend was present also for severe hematological toxicity during maintenance. In IBD, no significant effect of rs2413739 could be found on TPMT activity, however azathioprine effectiveness was reduced in patients carrying the T allele (linear mixed effect, p = 0.0058). In PBMC from healthy donors, a positive correlation between PACSIN2 and TPMT protein concentration could be detected (linear mixed effect, p = 0.045). These results support the role of PACSIN2 polymorphism on TPMT activity and mercaptopurine adverse effects in patients with ALL. Further evidence on PBMC and pediatric patients with IBD supports an association between PACSIN2 variants, TPMT activity, and thiopurines effects, even if more studies are needed since some of these effects may be tissue specific.
Collapse
|
23
|
Egawa J, Zemljic-Harpf A, Mandyam CD, Niesman IR, Lysenko LV, Kleschevnikov AM, Roth DM, Patel HH, Patel PM, Head BP. Neuron-Targeted Caveolin-1 Promotes Ultrastructural and Functional Hippocampal Synaptic Plasticity. Cereb Cortex 2019; 28:3255-3266. [PMID: 28981594 DOI: 10.1093/cercor/bhx196] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2017] [Indexed: 12/15/2022] Open
Abstract
A delicate interneuronal communication between pre- and postsynaptic membranes is critical for synaptic plasticity and the formation of memory. Evidence shows that membrane/lipid rafts (MLRs), plasma membrane microdomains enriched in cholesterol and sphingolipids, organize presynaptic proteins and postsynaptic receptors necessary for synaptic formation and signaling. MLRs establish a cell polarity that facilitates transduction of extracellular cues to the intracellular environment. Here we show that neuron-targeted overexpression of an MLR protein, caveolin-1 (SynCav1), in the adult mouse hippocampus increased the number of presynaptic vesicles per bouton, total excitatory type I glutamatergic synapses, number of same-dendrite multiple-synapse boutons, increased myelination, increased long-term potentiation, and increased MLR-localized N-methyl-d-aspartate receptor subunits (GluN1, GluN2A, and GluN2B). Immunogold electron microscopy revealed that Cav-1 localizes to both the pre- and postsynaptic membrane regions as well as in the synaptic cleft. These findings, which are consistent with a significant increase in ultrastructural and functional synaptic plasticity, provide a fundamental framework that underlies previously demonstrated improvements in learning and memory in adult and aged mice by SynCav1. Such observations suggest that Cav-1 and MLRs alter basic aspects of synapse biology that could serve as potential therapeutic targets to promote neuroplasticity and combat neurodegeneration in a number of neurological disorders.
Collapse
Affiliation(s)
- Junji Egawa
- Veterans Affairs San Diego Healthcare System, San Diego, CA, USA.,Department of Anesthesiology, School of Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Alice Zemljic-Harpf
- Veterans Affairs San Diego Healthcare System, San Diego, CA, USA.,Department of Anesthesiology, School of Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Chitra D Mandyam
- Veterans Affairs San Diego Healthcare System, San Diego, CA, USA.,Department of Anesthesiology, School of Medicine, University of California, San Diego, La Jolla, CA, USA
| | | | - Larisa V Lysenko
- Department of Neurosciences, University of California, San Diego, La Jolla, CA, USA
| | | | - David M Roth
- Veterans Affairs San Diego Healthcare System, San Diego, CA, USA.,Department of Anesthesiology, School of Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Hemal H Patel
- Veterans Affairs San Diego Healthcare System, San Diego, CA, USA.,Department of Anesthesiology, School of Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Piyush M Patel
- Veterans Affairs San Diego Healthcare System, San Diego, CA, USA.,Department of Anesthesiology, School of Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Brian P Head
- Veterans Affairs San Diego Healthcare System, San Diego, CA, USA.,Department of Anesthesiology, School of Medicine, University of California, San Diego, La Jolla, CA, USA
| |
Collapse
|
24
|
Sterk C, Gräber L, Schmitz HP, Heinisch JJ. Analysis of Functional Domains in Rho5, the Yeast Homolog of Human Rac1 GTPase, in Oxidative Stress Response. Int J Mol Sci 2019; 20:ijms20225550. [PMID: 31703278 PMCID: PMC6888954 DOI: 10.3390/ijms20225550] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Revised: 11/03/2019] [Accepted: 11/05/2019] [Indexed: 12/23/2022] Open
Abstract
The small GTPase Rho5 of Saccharomyces cerevisiae is required for proper regulation of different signaling pathways, which includes the response to cell wall, osmotic, nutrient, and oxidative stress. We here show that proper in vivo function and intracellular distribution of Rho5 depends on its hypervariable region at the carboxyterminal end, which includes the CAAX box for lipid modification, a preceding polybasic region (PBR) carrying a serine residue, and a 98 amino acid–specific insertion only present in Rho5 of S. cerevisiae but not in its human homolog Rac1. Results from trapping GFP-Rho5 variants to the mitochondrial surface suggest that the GTPase needs to be activated at the plasma membrane prior to its translocation to mitochondria in order to fulfil its role in oxidative stress response. These findings are supported by heterologous expression of a codon-optimized human RAC1 gene, which can only complement a yeast rho5 deletion in a chimeric fusion with RHO5 sequences that restore the correct spatiotemporal distribution of the encoded protein.
Collapse
|
25
|
Postema MM, Grega-Larson NE, Meenderink LM, Tyska MJ. PACSIN2-dependent apical endocytosis regulates the morphology of epithelial microvilli. Mol Biol Cell 2019; 30:2515-2526. [PMID: 31390291 PMCID: PMC6743356 DOI: 10.1091/mbc.e19-06-0352] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Apical microvilli are critical for the homeostasis of transporting epithelia, yet mechanisms that control the assembly and morphology of these protrusions remain poorly understood. Previous studies in intestinal epithelial cell lines suggested a role for the F-BAR domain protein PACSIN2 in normal microvillar assembly. Here we report the phenotype of PACSIN2 KO mice and provide evidence that through its role in promoting apical endocytosis, this molecule plays a role in controlling microvillar morphology. PACSIN2 KO enterocytes exhibit reduced numbers of microvilli and defects in the microvillar ultrastructure, with membranes lifting away from rootlets of core bundles. Dynamin2, a PACSIN2 binding partner, and other endocytic factors were also lost from their normal localization near microvillar rootlets. To determine whether loss of endocytic machinery could explain defects in microvillar morphology, we examined the impact of PACSIN2 KD and endocytosis inhibition on live intestinal epithelial cells. These assays revealed that when endocytic vesicle scission fails, tubules are pulled into the cytoplasm and this, in turn, leads to a membrane-lifting phenomenon reminiscent of that observed at PACSIN2 KO brush borders. These findings lead to a new model where inward forces generated by endocytic machinery on the plasma membrane control the membrane wrapping of cell surface protrusions.
Collapse
Affiliation(s)
- Meagan M Postema
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, University Medical Center, Nashville, TN 37232
| | - Nathan E Grega-Larson
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, University Medical Center, Nashville, TN 37232
| | - Leslie M Meenderink
- Division of Infectious Diseases, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232
| | - Matthew J Tyska
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, University Medical Center, Nashville, TN 37232
| |
Collapse
|
26
|
Lakshmanan M, Kok YJ, Lee AP, Kyriakopoulos S, Lim HL, Teo G, Poh SL, Tang WQ, Hong J, Tan AH, Bi X, Ho YS, Zhang P, Ng SK, Lee D. Multi‐omics profiling of CHO parental hosts reveals cell line‐specific variations in bioprocessing traits. Biotechnol Bioeng 2019; 116:2117-2129. [DOI: 10.1002/bit.27014] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Revised: 04/03/2019] [Accepted: 05/02/2019] [Indexed: 12/17/2022]
Affiliation(s)
- Meiyappan Lakshmanan
- Bioprocessing Technology InstituteAgency for Science, Technology and Research (A*STAR) Singapore
| | - Yee Jiun Kok
- Bioprocessing Technology InstituteAgency for Science, Technology and Research (A*STAR) Singapore
| | - Alison P. Lee
- Bioprocessing Technology InstituteAgency for Science, Technology and Research (A*STAR) Singapore
| | - Sarantos Kyriakopoulos
- Bioprocessing Technology InstituteAgency for Science, Technology and Research (A*STAR) Singapore
| | - Hsueh Lee Lim
- Bioprocessing Technology InstituteAgency for Science, Technology and Research (A*STAR) Singapore
| | - Gavin Teo
- Bioprocessing Technology InstituteAgency for Science, Technology and Research (A*STAR) Singapore
| | - Swan Li Poh
- Bioprocessing Technology InstituteAgency for Science, Technology and Research (A*STAR) Singapore
| | - Wen Qin Tang
- Bioprocessing Technology InstituteAgency for Science, Technology and Research (A*STAR) Singapore
| | - Jongkwang Hong
- Bioprocessing Technology InstituteAgency for Science, Technology and Research (A*STAR) Singapore
| | - Andy Hee‐Meng Tan
- Bioprocessing Technology InstituteAgency for Science, Technology and Research (A*STAR) Singapore
| | - Xuezhi Bi
- Bioprocessing Technology InstituteAgency for Science, Technology and Research (A*STAR) Singapore
| | - Ying Swan Ho
- Bioprocessing Technology InstituteAgency for Science, Technology and Research (A*STAR) Singapore
| | - Peiqing Zhang
- Bioprocessing Technology InstituteAgency for Science, Technology and Research (A*STAR) Singapore
| | - Say Kong Ng
- Bioprocessing Technology InstituteAgency for Science, Technology and Research (A*STAR) Singapore
| | - Dong‐Yup Lee
- Bioprocessing Technology InstituteAgency for Science, Technology and Research (A*STAR) Singapore
- School of Chemical EngineeringSungkyunkwan UniversitySuwon Republic of Korea
| |
Collapse
|
27
|
Mendoza-Topaz C, Yeow I, Riento K, Nichols BJ. BioID identifies proteins involved in the cell biology of caveolae. PLoS One 2018; 13:e0209856. [PMID: 30589899 PMCID: PMC6307745 DOI: 10.1371/journal.pone.0209856] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Accepted: 12/12/2018] [Indexed: 01/20/2023] Open
Abstract
The mechanisms controlling the abundance and sub-cellular distribution of caveolae are not well described. A first step towards determining such mechanisms would be identification of relevant proteins that interact with known components of caveolae. Here, we applied proximity biotinylation (BioID) to identify a list of proteins that may interact with the caveolar protein cavin1. Screening of these candidates using siRNA to reduce their expression revealed that one of them, CSDE1, regulates the levels of mRNAs and protein expression for multiple components of caveolae. A second candidate, CD2AP, co-precipitated with cavin1. Caveolar proteins were observed in characteristic and previously un-described linear arrays adjacent to cell-cell junctions in both MDCK cells, and in HeLa cells overexpressing an active form of the small GTPase Rac1. CD2AP was required for the recruitment of caveolar proteins to these linear arrays. We conclude that BioID will be useful in identification of new proteins involved in the cell biology of caveolae, and that interaction between CD2AP and cavin1 may have an important role in regulating the sub-cellular distribution of caveolae.
Collapse
Affiliation(s)
| | - I. Yeow
- MRC Laboratory of Molecular Biology, Cambridge, United Kingdom
| | - K. Riento
- MRC Laboratory of Molecular Biology, Cambridge, United Kingdom
| | - B. J. Nichols
- MRC Laboratory of Molecular Biology, Cambridge, United Kingdom
- * E-mail:
| |
Collapse
|
28
|
BAR domain proteins-a linkage between cellular membranes, signaling pathways, and the actin cytoskeleton. Biophys Rev 2018; 10:1587-1604. [PMID: 30456600 DOI: 10.1007/s12551-018-0467-7] [Citation(s) in RCA: 90] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2018] [Accepted: 10/17/2018] [Indexed: 12/23/2022] Open
Abstract
Actin filament assembly typically occurs in association with cellular membranes. A large number of proteins sit at the interface between actin networks and membranes, playing diverse roles such as initiation of actin polymerization, modulation of membrane curvature, and signaling. Bin/Amphiphysin/Rvs (BAR) domain proteins have been implicated in all of these functions. The BAR domain family of proteins comprises a diverse group of multi-functional effectors, characterized by their modular architecture. In addition to the membrane-curvature sensing/inducing BAR domain module, which also mediates antiparallel dimerization, most contain auxiliary domains implicated in protein-protein and/or protein-membrane interactions, including SH3, PX, PH, RhoGEF, and RhoGAP domains. The shape of the BAR domain itself varies, resulting in three major subfamilies: the classical crescent-shaped BAR, the more extended and less curved F-BAR, and the inverse curvature I-BAR subfamilies. Most members of this family have been implicated in cellular functions that require dynamic remodeling of the actin cytoskeleton, such as endocytosis, organelle trafficking, cell motility, and T-tubule biogenesis in muscle cells. Here, we review the structure and function of mammalian BAR domain proteins and the many ways in which they are interconnected with the actin cytoskeleton.
Collapse
|
29
|
Lim SZ, Chua EW. Revisiting the Role of Thiopurines in Inflammatory Bowel Disease Through Pharmacogenomics and Use of Novel Methods for Therapeutic Drug Monitoring. Front Pharmacol 2018; 9:1107. [PMID: 30349479 PMCID: PMC6186994 DOI: 10.3389/fphar.2018.01107] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Accepted: 09/10/2018] [Indexed: 12/12/2022] Open
Abstract
Azathioprine and 6-mercaptopurine, often referred to as thiopurine compounds, are commonly used in the management of inflammatory bowel disease. However, patients receiving these drugs are prone to developing adverse drug reactions or therapeutic resistance. Achieving predefined levels of two major thiopurine metabolites, 6-thioguanine nucleotides and 6-methylmercaptopurine, is a long-standing clinical practice in ensuring therapeutic efficacy; however, their correlation with treatment response is sometimes unclear. Various genetic markers have also been used to aid the identification of patients who are thiopurine-sensitive or refractory. The recent discovery of novel Asian-specific DNA variants, namely those in the NUDT15 gene, and their link to thiopurine toxicity, have led clinicians and scientists to revisit the utility of Caucasian biomarkers for Asian individuals with inflammatory bowel disease. In this review, we explore the limitations associated with the current methods used for therapeutic monitoring of thiopurine metabolites and how the recent discovery of ethnicity-specific genetic markers can complement thiopurine metabolites measurement in formulating a strategy for more accurate prediction of thiopurine response. We also discuss the challenges in thiopurine therapy, alongside the current strategies used in patients with reduced thiopurine response. The review is concluded with suggestions for future work aiming at using a more comprehensive approach to optimize the efficacy of thiopurine compounds in inflammatory bowel disease.
Collapse
Affiliation(s)
| | - Eng Wee Chua
- Faculty of Pharmacy, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| |
Collapse
|
30
|
Aspenström P. BAR Domain Proteins Regulate Rho GTPase Signaling. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1111:33-53. [PMID: 30151649 DOI: 10.1007/5584_2018_259] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The Bin-Amphiphysin-Rvs (BAR) domain is a membrane lipid binding domain present in a wide variety of proteins, often proteins with a role in Rho-regulated signaling pathways. BAR domains do not only confer binding to lipid bilayers, they also possess a membrane sculpturing ability and thereby directly control the topology of biomembranes. BAR domain-containing proteins participate in a plethora of physiological processes but the common denominator is their capacity to link membrane dynamics to actin dynamics and thereby integrate processes such as endocytosis, exocytosis, vesicle trafficking, cell morphogenesis and cell migration. The Rho family of small GTPases constitutes an important bridging theme for many BAR domain-containing proteins. This review article will focus predominantly on the role of BAR proteins as regulators or effectors of Rho GTPases and it will only briefly discuss the structural and biophysical function of the BAR domains.
Collapse
Affiliation(s)
- Pontus Aspenström
- Department of Microbiology, and Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
31
|
Mani M, Lee UH, Yoon NA, Yoon EH, Lee BJ, Cho WJ, Park JW. Developmentally regulated GTP-binding protein 2 is required for stabilization of Rac1-positive membrane tubules. Biochem Biophys Res Commun 2017; 493:758-764. [PMID: 28865956 DOI: 10.1016/j.bbrc.2017.08.110] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Accepted: 08/27/2017] [Indexed: 01/07/2023]
Abstract
Previously we have reported that developmentally regulated GTP-binding protein 2 (DRG2) localizes on Rab5 endosomes and plays an important role in transferrin (Tfn) recycling. We here identified DRG2 as a key regulator of membrane tubule stability. At 30 min after Tfn treatment, DRG2 localized to membrane tubules which were enriched with phosphatidylinositol 4-monophosphate [PI(4)P] and did not contain Rab5. DRG2 interacted with Rac1 more strongly with GTP-bound Rac1 and tubular localization of DRG2 depended on Rac1 activity. DRG2 depletion led to destabilization of membrane tubules, while ectopic expression of DRG2 rescued the stability of the membrane tubules in DRG2-depleted cells. Our results reveal a novel mechanism for regulation of membrane tubule stability mediated by DRG2.
Collapse
Affiliation(s)
- Muralidharan Mani
- Department of Biological Sciences, University of Ulsan, Ulsan 680-749, South Korea
| | - Unn Hwa Lee
- Department of Biological Sciences, University of Ulsan, Ulsan 680-749, South Korea
| | - Nal Ae Yoon
- Department of Biological Sciences, University of Ulsan, Ulsan 680-749, South Korea
| | - Eun Hye Yoon
- Department of Biological Sciences, University of Ulsan, Ulsan 680-749, South Korea
| | - Byung Ju Lee
- Department of Biological Sciences, University of Ulsan, Ulsan 680-749, South Korea
| | - Wha Ja Cho
- Metainflammation Research Center, University of Ulsan, Ulsan 680-749, South Korea
| | - Jeong Woo Park
- Department of Biological Sciences, University of Ulsan, Ulsan 680-749, South Korea.
| |
Collapse
|
32
|
Semmler J, Kormann J, Srinivasan SP, Köster A, Sälzer D, Reppel M, Hescheler J, Plomann M, Nguemo F. Pacsin 2 is required for the maintenance of a normal cardiac function in the developing mouse heart. Pharmacol Res 2017; 128:200-210. [PMID: 29107716 DOI: 10.1016/j.phrs.2017.10.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2016] [Revised: 06/26/2017] [Accepted: 10/15/2017] [Indexed: 11/27/2022]
Abstract
The Pacsin proteins (Pacsin 1, 2 and 3) play an important role in intracellular trafficking and thereby signal transduction in many cells types. This study was designed to examine the role of Pacsin 2 in cardiac development and function. We investigated the development and electrophysiological properties of Pacsin 2 knockout (P2KO) hearts and single cardiomyocytes isolated from 11.5 and 15.5days old fetal mice. Immunofluorescence experiments confirmed the lack of Pacsin 2 protein expression in P2KO cardiac myocytes in comparison to wildtype (WT). Western blotting demonstrates low expression levels of connexin 43 and T-box 3 proteins in P2KO compared to wildtype (WT). Electrophysiology measurements including online Multi-Electrode Array (MEA) based field potential (FP) recordings on isolated whole heart of P2KO mice showed a prolonged AV-conduction time. Patch clamp measurements of P2KO cardiomyocytes revealed differences in action potential (AP) parameters and decreased pacemaker funny channel (If), as well as L-type Ca2+ channel (ICaL), and sodium channel (INa). These findings demonstrate that Pacsin 2 is necessary for cardiac development and function in mouse embryos, which will enhance our knowledge to better understand the genesis of cardiovascular diseases.
Collapse
Affiliation(s)
- Judith Semmler
- Institute of Neurophysiology, University of Cologne, 50931 Cologne, Germany
| | - Jan Kormann
- Institute of Biochemistry, University of Cologne, 50931 Cologne, Germany
| | | | - Annette Köster
- Institute of Neurophysiology, University of Cologne, 50931 Cologne, Germany
| | - Daniel Sälzer
- Institute of Biochemistry, University of Cologne, 50931 Cologne, Germany
| | - Michael Reppel
- Institute of Neurophysiology, University of Cologne, 50931 Cologne, Germany; Department of Cardiology, University of Lübeck, Lübeck, Germany
| | - Jürgen Hescheler
- Institute of Neurophysiology, University of Cologne, 50931 Cologne, Germany
| | - Markus Plomann
- Institute of Biochemistry, University of Cologne, 50931 Cologne, Germany
| | - Filomain Nguemo
- Institute of Neurophysiology, University of Cologne, 50931 Cologne, Germany.
| |
Collapse
|
33
|
Soriano-Castell D, Chavero A, Rentero C, Bosch M, Vidal-Quadras M, Pol A, Enrich C, Tebar F. ROCK1 is a novel Rac1 effector to regulate tubular endocytic membrane formation during clathrin-independent endocytosis. Sci Rep 2017; 7:6866. [PMID: 28761175 PMCID: PMC5537229 DOI: 10.1038/s41598-017-07130-x] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2016] [Accepted: 06/23/2017] [Indexed: 01/10/2023] Open
Abstract
Clathrin-dependent and -independent pathways contribute for β1-integrin endocytosis. This study defines a tubular membrane clathrin-independent endocytic network, induced with the calmodulin inhibitor W13, for β1-integrin internalization. This pathway is dependent on increased phosphatidylinositol 4,5-bisphosphate (PI(4,5)P2) levels and dynamin activity at the plasma membrane. Exogenous addition of PI(4,5)P2 or phosphatidylinositol-4-phosphate 5-kinase (PIP5K) expression mimicked W13-generated-tubules which are inhibited by active Rac1. Therefore, the molecular mechanisms downstream of Rac1, that controls this plasma membrane tubulation, were analyzed biochemically and by the expression of different Rac1 mutants. The results indicate that phospholipase C and ROCK1 are the main Rac1 effectors that impair plasma membrane invagination and tubule formation, essentially by decreasing PI(4,5)P2 levels and promoting cortical actomyosin assembly respectively. Interestingly, among the plethora of proteins that participate in membrane remodeling, this study revealed that ROCK1, the well-known downstream RhoA effector, has an important role in Rac1 regulation of actomyosin at the cell cortex. This study provides new insights into Rac1 functioning on plasma membrane dynamics combining phosphatidylinositides and cytoskeleton regulation.
Collapse
Affiliation(s)
- David Soriano-Castell
- Departament de Biomedicina, Unitat de Biologia Cel·lular, Centre de Recerca Biomèdica CELLEX, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Facultat de Medicina, Universitat de Barcelona, Casanova 143, 08036, Barcelona, Spain
| | - Albert Chavero
- Departament de Biomedicina, Unitat de Biologia Cel·lular, Centre de Recerca Biomèdica CELLEX, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Facultat de Medicina, Universitat de Barcelona, Casanova 143, 08036, Barcelona, Spain
| | - Carles Rentero
- Departament de Biomedicina, Unitat de Biologia Cel·lular, Centre de Recerca Biomèdica CELLEX, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Facultat de Medicina, Universitat de Barcelona, Casanova 143, 08036, Barcelona, Spain
| | - Marta Bosch
- Departament de Biomedicina, Unitat de Biologia Cel·lular, Centre de Recerca Biomèdica CELLEX, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Facultat de Medicina, Universitat de Barcelona, Casanova 143, 08036, Barcelona, Spain
| | - Maite Vidal-Quadras
- Departament de Biomedicina, Unitat de Biologia Cel·lular, Centre de Recerca Biomèdica CELLEX, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Facultat de Medicina, Universitat de Barcelona, Casanova 143, 08036, Barcelona, Spain
| | - Albert Pol
- Departament de Biomedicina, Unitat de Biologia Cel·lular, Centre de Recerca Biomèdica CELLEX, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Facultat de Medicina, Universitat de Barcelona, Casanova 143, 08036, Barcelona, Spain.,Institució Catalana de Recerca i Estudis Avançats (ICREA), 08010, Barcelona, Spain
| | - Carlos Enrich
- Departament de Biomedicina, Unitat de Biologia Cel·lular, Centre de Recerca Biomèdica CELLEX, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Facultat de Medicina, Universitat de Barcelona, Casanova 143, 08036, Barcelona, Spain
| | - Francesc Tebar
- Departament de Biomedicina, Unitat de Biologia Cel·lular, Centre de Recerca Biomèdica CELLEX, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Facultat de Medicina, Universitat de Barcelona, Casanova 143, 08036, Barcelona, Spain.
| |
Collapse
|
34
|
Pearn ML, Niesman IR, Egawa J, Sawada A, Almenar-Queralt A, Shah SB, Duckworth JL, Head BP. Pathophysiology Associated with Traumatic Brain Injury: Current Treatments and Potential Novel Therapeutics. Cell Mol Neurobiol 2017; 37:571-585. [PMID: 27383839 DOI: 10.1007/s10571-016-0400-1] [Citation(s) in RCA: 201] [Impact Index Per Article: 28.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2016] [Accepted: 06/24/2016] [Indexed: 12/20/2022]
Abstract
Traumatic brain injury (TBI) is one of the leading causes of death of young people in the developed world. In the United States alone, 1.7 million traumatic events occur annually accounting for 50,000 deaths. The etiology of TBI includes traffic accidents, falls, gunshot wounds, sports, and combat-related events. TBI severity ranges from mild to severe. TBI can induce subtle changes in molecular signaling, alterations in cellular structure and function, and/or primary tissue injury, such as contusion, hemorrhage, and diffuse axonal injury. TBI results in blood-brain barrier (BBB) damage and leakage, which allows for increased extravasation of immune cells (i.e., increased neuroinflammation). BBB dysfunction and impaired homeostasis contribute to secondary injury that occurs from hours to days to months after the initial trauma. This delayed nature of the secondary injury suggests a potential therapeutic window. The focus of this article is on the (1) pathophysiology of TBI and (2) potential therapies that include biologics (stem cells, gene therapy, peptides), pharmacological (anti-inflammatory, antiepileptic, progrowth), and noninvasive (exercise, transcranial magnetic stimulation). In final, the review briefly discusses membrane/lipid rafts (MLR) and the MLR-associated protein caveolin (Cav). Interventions that increase Cav-1, MLR formation, and MLR recruitment of growth-promoting signaling components may augment the efficacy of pharmacologic agents or already existing endogenous neurotransmitters and neurotrophins that converge upon progrowth signaling cascades resulting in improved neuronal function after injury.
Collapse
Affiliation(s)
- Matthew L Pearn
- Department of Anesthesiology, Veterans Affairs San Diego Healthcare System, VA Medical Center 125, University of California, 3350 La Jolla Village Drive, San Diego, CA, 92161-5085, USA
- Department of Anesthesiology, School of Medicine, University of California, La Jolla, San Diego, CA, 92093, USA
| | - Ingrid R Niesman
- Department of Cellular and Molecular Medicine, University of California, La Jolla, San Diego, CA, 92093, USA
- Sanford Consortium for Regenerative Medicine, La Jolla, San Diego, CA, 92037, USA
| | - Junji Egawa
- Department of Anesthesiology, Veterans Affairs San Diego Healthcare System, VA Medical Center 125, University of California, 3350 La Jolla Village Drive, San Diego, CA, 92161-5085, USA
- Department of Anesthesiology, School of Medicine, University of California, La Jolla, San Diego, CA, 92093, USA
| | - Atsushi Sawada
- Department of Anesthesiology, Veterans Affairs San Diego Healthcare System, VA Medical Center 125, University of California, 3350 La Jolla Village Drive, San Diego, CA, 92161-5085, USA
- Department of Anesthesiology, School of Medicine, University of California, La Jolla, San Diego, CA, 92093, USA
| | - Angels Almenar-Queralt
- Department of Cellular and Molecular Medicine, University of California, La Jolla, San Diego, CA, 92093, USA
- Sanford Consortium for Regenerative Medicine, La Jolla, San Diego, CA, 92037, USA
| | - Sameer B Shah
- UCSD Departments of Orthopaedic Surgery and Bioengineering, University of California, La Jolla, San Diego, CA, 92093, USA
| | - Josh L Duckworth
- Department of Neurology, F. Edward Hébert School of Medicine, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, Bethesda, MD, 20814, USA
| | - Brian P Head
- Department of Anesthesiology, Veterans Affairs San Diego Healthcare System, VA Medical Center 125, University of California, 3350 La Jolla Village Drive, San Diego, CA, 92161-5085, USA.
- Department of Anesthesiology, School of Medicine, University of California, La Jolla, San Diego, CA, 92093, USA.
| |
Collapse
|
35
|
Mandyam CD, Schilling JM, Cui W, Egawa J, Niesman IR, Kellerhals SE, Staples MC, Busija AR, Risbrough VB, Posadas E, Grogman GC, Chang JW, Roth DM, Patel PM, Patel HH, Head BP. Neuron-Targeted Caveolin-1 Improves Molecular Signaling, Plasticity, and Behavior Dependent on the Hippocampus in Adult and Aged Mice. Biol Psychiatry 2017; 81:101-110. [PMID: 26592463 PMCID: PMC4826329 DOI: 10.1016/j.biopsych.2015.09.020] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2015] [Revised: 09/29/2015] [Accepted: 09/30/2015] [Indexed: 01/19/2023]
Abstract
BACKGROUND Studies in vitro demonstrate that neuronal membrane/lipid rafts (MLRs) establish cell polarity by clustering progrowth receptors and tethering cytoskeletal machinery necessary for neuronal sprouting. However, the effect of MLR and MLR-associated proteins on neuronal aging is unknown. METHODS Here, we assessed the impact of neuron-targeted overexpression of an MLR scaffold protein, caveolin-1 (Cav-1) (via a synapsin promoter, SynCav1), in the hippocampus in vivo in adult (6-month-old) and aged (20-month-old) mice on biochemical, morphologic, and behavioral changes. RESULTS SynCav1 resulted in increased expression of Cav-1, MLRs, and MLR-localization of Cav-1 and tropomyosin-related kinase B receptor independent of age and time post gene transfer. Cav-1 overexpression in adult mice enhanced dendritic arborization within the apical dendrites of hippocampal cornu ammonis 1 and granule cell neurons, effects that were also observed in aged mice, albeit to a lesser extent, indicating preserved impact of Cav-1 on structural plasticity of hippocampal neurons with age. Cav-1 overexpression enhanced contextual fear memory in adult and aged mice demonstrating improved hippocampal function. CONCLUSIONS Neuron-targeted overexpression of Cav-1 in the adult and aged hippocampus enhances functional MLRs with corresponding roles in cell signaling and protein trafficking. The resultant structural alterations in hippocampal neurons in vivo are associated with improvements in hippocampal-dependent learning and memory. Our findings suggest Cav-1 as a novel therapeutic strategy in disorders involving impaired hippocampal function.
Collapse
Affiliation(s)
- Chitra D. Mandyam
- Skaggs School of Pharmacy and Pharmaceutical Sciences, UCSD,Committee on the Neurobiology of Addictive Disorders, TSRI
| | - Jan M. Schilling
- Veterans Affairs San Diego Healthcare System,Department of Anesthesiology, UCSD
| | - Weihua Cui
- Veterans Affairs San Diego Healthcare System,Department of Anesthesiology, UCSD,Department of Anesthesiology, Beijing Tiantan Hospital, Capital Medical University
| | - Junji Egawa
- Veterans Affairs San Diego Healthcare System,Department of Anesthesiology, UCSD
| | - Ingrid R. Niesman
- Department of Cellular and Molecular Medicine, UCSD, Sanford Consortium for Regenerative Medicine
| | - Sarah E. Kellerhals
- Veterans Affairs San Diego Healthcare System,Department of Anesthesiology, UCSD
| | | | - Anna R. Busija
- Veterans Affairs San Diego Healthcare System,Department of Anesthesiology, UCSD
| | | | - Edmund Posadas
- Veterans Affairs San Diego Healthcare System,Department of Anesthesiology, UCSD
| | - Grace C. Grogman
- Veterans Affairs San Diego Healthcare System,Department of Anesthesiology, UCSD
| | - Jamie W. Chang
- Veterans Affairs San Diego Healthcare System,Department of Anesthesiology, UCSD
| | - David M. Roth
- Veterans Affairs San Diego Healthcare System,Department of Anesthesiology, UCSD
| | - Piyush M. Patel
- Veterans Affairs San Diego Healthcare System,Department of Anesthesiology, UCSD
| | - Hemal H. Patel
- Veterans Affairs San Diego Healthcare System,Department of Anesthesiology, UCSD
| | - Brian P. Head
- Veterans Affairs San Diego Healthcare System,Department of Anesthesiology, UCSD,Corresponding Author: Brian P. Head, Department of Anesthesiology, University of California San Diego, VASDHS (9125), 3350 La Jolla Village Dr., San Diego, CA 92161, USA.
| |
Collapse
|
36
|
Chandrasekaran R, Kenworthy AK, Lacy DB. Clostridium difficile Toxin A Undergoes Clathrin-Independent, PACSIN2-Dependent Endocytosis. PLoS Pathog 2016; 12:e1006070. [PMID: 27942025 PMCID: PMC5152916 DOI: 10.1371/journal.ppat.1006070] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2016] [Accepted: 11/17/2016] [Indexed: 12/18/2022] Open
Abstract
Clostridium difficile infection affects a significant number of hospitalized patients in the United States. Two homologous exotoxins, TcdA and TcdB, are the major virulence factors in C. difficile pathogenesis. The toxins are glucosyltransferases that inactivate Rho family-GTPases to disrupt host cellular function and cause fluid secretion, inflammation, and cell death. Toxicity depends on receptor binding and subsequent endocytosis. TcdB has been shown to enter cells by clathrin-dependent endocytosis, but the mechanism of TcdA uptake is still unclear. Here, we utilize a combination of RNAi-based knockdown, pharmacological inhibition, and cell imaging approaches to investigate the endocytic mechanism(s) that contribute to TcdA uptake and subsequent cytopathic and cytotoxic effects. We show that TcdA uptake and cellular intoxication is dynamin-dependent but does not involve clathrin- or caveolae-mediated endocytosis. Confocal microscopy using fluorescently labeled TcdA shows significant colocalization of the toxin with PACSIN2-positive structures in cells during entry. Disruption of PACSIN2 function by RNAi-based knockdown approaches inhibits TcdA uptake and toxin-induced downstream effects in cells indicating that TcdA entry is PACSIN2-dependent. We conclude that TcdA and TcdB utilize distinct endocytic mechanisms to intoxicate host cells. Clostridium difficile is a bacterial pathogen that causes nearly half a million infections each year in the United States. It infects the human colon and causes diarrhea, colitis and, in some cases, death. C. difficile infection is mediated by the action of two large homologous toxins, TcdA and TcdB. Disruption of host cell function by these toxins requires entry into cells. There are multiple ways for pathogens and virulence factors such as viruses and toxins to enter host cells. The entry mechanism is often directed by a cell surface receptor and can impact the trafficking and virulence properties of the pathogenic factor. Investigating the internalization strategy can provide critical insight into the mechanism of action for specific pathogens and virulence factors. In our current study, we sought to determine the strategy utilized by TcdA to enter host cells. We show that TcdA uptake occurs by a clathrin- and caveolae-independent endocytic mechanism that is mediated by PACSIN2 and dynamin. We also show that TcdA and TcdB can utilize different routes of entry, which may have implications regarding their cytotoxic mechanisms. In summary, our results provide new insights into the mechanism of cellular intoxication by TcdA and the role of PACSIN2 in endocytosis.
Collapse
Affiliation(s)
- Ramyavardhanee Chandrasekaran
- Department of Pathology, Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, TN, United States of America
| | - Anne K. Kenworthy
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN, United States of America
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN, United States of America
- Epithelial Biology Program, Vanderbilt University School of Medicine, Nashville, TN, United States of America
| | - D. Borden Lacy
- Department of Pathology, Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, TN, United States of America
- Epithelial Biology Program, Vanderbilt University School of Medicine, Nashville, TN, United States of America
- The Veterans Affairs Tennessee Valley Healthcare System, Nashville, TN, United States of America
- * E-mail:
| |
Collapse
|
37
|
Characterization of Novel Molecular Mechanisms Favoring Rac1 Membrane Translocation. PLoS One 2016; 11:e0166715. [PMID: 27835684 PMCID: PMC5105943 DOI: 10.1371/journal.pone.0166715] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2016] [Accepted: 11/02/2016] [Indexed: 11/19/2022] Open
Abstract
The Rac1 GTPase plays key roles in cytoskeletal organization, cell motility and a variety of physiological and disease-linked responses. Wild type Rac1 signaling entails dissociation of the GTPase from cytosolic Rac1-Rho GDP dissociation inhibitor (GDI) complexes, translocation to membranes, activation by exchange factors, effector binding, and activation of downstream signaling cascades. Out of those steps, membrane translocation is the less understood. Using transfections of a expression cDNA library in cells expressing a Rac1 bioreporter, we previously identified a cytoskeletal feedback loop nucleated by the F-actin binding protein coronin 1A (Coro1A) that promotes Rac1 translocation to the plasma membrane by facilitating the Pak-dependent dissociation of Rac1-Rho GDI complexes. This screening identified other potential regulators of this process, including WDR26, basigin, and TMEM8A. Here, we show that WDR26 promotes Rac1 translocation following a Coro1A-like and Coro1A-dependent mechanism. By contrast, basigin and TMEM8A stabilize Rac1 at the plasma membrane by inhibiting the internalization of caveolin-rich membrane subdomains. This latter pathway is F-actin-dependent but Coro1A-, Pak- and Rho GDI-independent.
Collapse
|
38
|
Senju Y, Suetsugu S. Possible regulation of caveolar endocytosis and flattening by phosphorylation of F-BAR domain protein PACSIN2/Syndapin II. BIOARCHITECTURE 2016; 5:70-7. [PMID: 26745030 PMCID: PMC4832444 DOI: 10.1080/19490992.2015.1128604] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
ABSTRACT. Caveolae are flask-shaped invaginations of the plasma membrane. The BAR domain proteins form crescent-shaped dimers, and their oligomeric filaments are considered to form spirals at the necks of invaginations, such as clathrin-coated pits and caveolae. PACSIN2/Syndapin II is one of the BAR domain-containing proteins, and is localized at the necks of caveolae. PACSIN2 is thought to function in the scission and stabilization of caveolae, through binding to dynamin-2 and EHD2, respectively. These two functions are considered to be switched by PACSIN2 phosphorylation by protein kinase C (PKC) upon hypotonic stress and sheer stress. The phosphorylation decreases the membrane binding affinity of PACSIN2, leading to its removal from caveolae. The removal of the putative oligomeric spiral of PACSIN2 from caveolar membrane invaginations could lead to the deformation of caveolae. Indeed, PACSIN2 removal from caveolae is accompanied by the recruitment of dynamin-2, suggesting that the removal provides space for the function of dynamin-2. Otherwise, the removal of PACSIN2 decreases the stability of caveolae, which could result in the flattening of caveolae. In contrast, an increase in the amount of EHD2 restored caveolar stability. Therefore, PACSIN2 at caveolae stabilizes caveolae, but its removal by phosphorylation could induce both caveolar endocytosis and flattening.
Collapse
Affiliation(s)
- Yosuke Senju
- a Institute of Biotechnology; University of Helsinki ; Helsinki , Finland
| | - Shiro Suetsugu
- b Laboratory of Molecular Medicine and Cell Biology; Graduate School of Biosciences; Nara Institute of Science and Technology ; Ikoma , Japan
| |
Collapse
|
39
|
Smid A, Karas-Kuzelicki N, Jazbec J, Mlinaric-Rascan I. PACSIN2 polymorphism is associated with thiopurine-induced hematological toxicity in children with acute lymphoblastic leukaemia undergoing maintenance therapy. Sci Rep 2016; 6:30244. [PMID: 27452984 PMCID: PMC4958958 DOI: 10.1038/srep30244] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2016] [Accepted: 07/01/2016] [Indexed: 02/07/2023] Open
Abstract
Adequate maintenance therapy for childhood acute lymphoblastic leukemia (ALL), with 6-mercaptopurine as an essential component, is necessary for retaining durable remission. Interruptions or discontinuations of the therapy due to drug-related toxicities, which can be life threatening, may result in an increased risk of relapse. In this retrospective study including 305 paediatric ALL patients undergoing maintenance therapy, we systematically investigated the individual and combined effects of genetic variants of folate pathway enzymes, as well as of polymorphisms in PACSIN2 and ITPA, on drug-induced toxicities by applying a multi-analytical approach including logistic regression (LR), classification and regression tree (CART) and generalized multifactor dimensionality reduction (GMDR). In addition to the TPMT genotype, confirmed to be a major determinant of drug related toxicities, we identified the PACSIN2 rs2413739TT genotype as being a significant risk factor for 6-MP-induced toxicity in wild-type TPMT patients. A gene-gene interaction between MTRR (rs1801394) and MTHFR (rs1801133) was detected by GMDR and proved to have an independent effect on the risk of stomatitis, as shown by LR analysis. To our knowledge, this is the first study showing PACSIN2 genotype association with hematological toxicity in ALL patients undergoing maintenance therapy.
Collapse
Affiliation(s)
- Alenka Smid
- Faculty of Pharmacy, University of Ljubljana, Ljubljana, Slovenia
| | | | - Janez Jazbec
- University Children's Hospital, University Medical Centre Ljubljana, Ljubljana, Slovenia
| | | |
Collapse
|
40
|
Dorland YL, Malinova TS, van Stalborch AMD, Grieve AG, van Geemen D, Jansen NS, de Kreuk BJ, Nawaz K, Kole J, Geerts D, Musters RJP, de Rooij J, Hordijk PL, Huveneers S. The F-BAR protein pacsin2 inhibits asymmetric VE-cadherin internalization from tensile adherens junctions. Nat Commun 2016; 7:12210. [PMID: 27417273 PMCID: PMC4947187 DOI: 10.1038/ncomms12210] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2015] [Accepted: 06/10/2016] [Indexed: 12/14/2022] Open
Abstract
Vascular homoeostasis, development and disease critically depend on the regulation of endothelial cell-cell junctions. Here we uncover a new role for the F-BAR protein pacsin2 in the control of VE-cadherin-based endothelial adhesion. Pacsin2 concentrates at focal adherens junctions (FAJs) that are experiencing unbalanced actomyosin-based pulling. FAJs move in response to differences in local cytoskeletal geometry and pacsin2 is recruited consistently to the trailing end of fast-moving FAJs via a mechanism that requires an intact F-BAR domain. Photoconversion, photobleaching, immunofluorescence and super-resolution microscopy reveal polarized dynamics, and organization of junctional proteins between the front of FAJs and their trailing ends. Interestingly, pacsin2 recruitment inhibits internalization of the VE-cadherin complex from FAJ trailing ends and is important for endothelial monolayer integrity. Together, these findings reveal a novel junction protective mechanism during polarized trafficking of VE-cadherin, which supports barrier maintenance within dynamic endothelial tissue.
Collapse
Affiliation(s)
- Yvonne L Dorland
- Department of Molecular Cell Biology, Sanquin Research and Landsteiner Laboratory, University of Amsterdam, Amsterdam 1066 CX, The Netherlands
| | - Tsveta S Malinova
- Department of Medical Biochemistry, Academic Medical Center, University of Amsterdam, Amsterdam 1105 AZ, The Netherlands
| | - Anne-Marieke D van Stalborch
- Department of Molecular Cell Biology, Sanquin Research and Landsteiner Laboratory, University of Amsterdam, Amsterdam 1066 CX, The Netherlands
| | - Adam G Grieve
- Hubrecht Institute and University Medical Center Utrecht, Utrecht 3584 CT, The Netherlands
| | - Daphne van Geemen
- Department of Molecular Cell Biology, Sanquin Research and Landsteiner Laboratory, University of Amsterdam, Amsterdam 1066 CX, The Netherlands
| | - Nicolette S Jansen
- Department of Molecular Cell Biology, Sanquin Research and Landsteiner Laboratory, University of Amsterdam, Amsterdam 1066 CX, The Netherlands
| | - Bart-Jan de Kreuk
- Department of Medicine, University of California, San Diego, California 92093, USA
| | - Kalim Nawaz
- Department of Molecular Cell Biology, Sanquin Research and Landsteiner Laboratory, University of Amsterdam, Amsterdam 1066 CX, The Netherlands
| | - Jeroen Kole
- Department of Physiology, VU University Medical Center, Amsterdam 1081 HV, The Netherlands
| | - Dirk Geerts
- Department of Pediatric Oncology/Hematology, Erasmus University Medical Center, Rotterdam 3015 GE, The Netherlands
| | - René J P Musters
- Department of Physiology, VU University Medical Center, Amsterdam 1081 HV, The Netherlands
| | - Johan de Rooij
- Department of Molecular Cancer Research, Center for Molecular Medicine, University Medical Center Utrecht, Utrecht 3584 CG, The Netherlands
| | - Peter L Hordijk
- Department of Physiology, VU University Medical Center, Amsterdam 1081 HV, The Netherlands
| | - Stephan Huveneers
- Department of Molecular Cell Biology, Sanquin Research and Landsteiner Laboratory, University of Amsterdam, Amsterdam 1066 CX, The Netherlands.,Department of Medical Biochemistry, Academic Medical Center, University of Amsterdam, Amsterdam 1105 AZ, The Netherlands
| |
Collapse
|
41
|
Sherlekar A, Rikhy R. Syndapin promotes pseudocleavage furrow formation by actin organization in the syncytial Drosophila embryo. Mol Biol Cell 2016; 27:2064-79. [PMID: 27146115 PMCID: PMC4927280 DOI: 10.1091/mbc.e15-09-0656] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2015] [Accepted: 04/26/2016] [Indexed: 12/03/2022] Open
Abstract
F-BAR domain–containing proteins link the actin cytoskeleton to the membrane during membrane remodeling. Syndapin associates with the pseudocleavage furrow membrane and is essential for furrow morphology, actin organization, and extension downstream of initiation factor RhoGEF2. Coordinated membrane and cytoskeletal remodeling activities are required for membrane extension in processes such as cytokinesis and syncytial nuclear division cycles in Drosophila. Pseudocleavage furrow membranes in the syncytial Drosophila blastoderm embryo show rapid extension and retraction regulated by actin-remodeling proteins. The F-BAR domain protein Syndapin (Synd) is involved in membrane tubulation, endocytosis, and, uniquely, in F-actin stability. Here we report a role for Synd in actin-regulated pseudocleavage furrow formation. Synd localized to these furrows, and its loss resulted in short, disorganized furrows. Synd presence was important for the recruitment of the septin Peanut and distribution of Diaphanous and F-actin at furrows. Synd and Peanut were both absent in furrow-initiation mutants of RhoGEF2 and Diaphanous and in furrow-progression mutants of Anillin. Synd overexpression in rhogef2 mutants reversed its furrow-extension phenotypes, Peanut and Diaphanous recruitment, and F-actin organization. We conclude that Synd plays an important role in pseudocleavage furrow extension, and this role is also likely to be crucial in cleavage furrow formation during cell division.
Collapse
Affiliation(s)
- Aparna Sherlekar
- Biology, Indian Institute of Science Education and Research, Pashan, Pune 411008, India
| | - Richa Rikhy
- Biology, Indian Institute of Science Education and Research, Pashan, Pune 411008, India
| |
Collapse
|
42
|
Wilson C, Saunter CD, Girkin JM, McCarron JG. Pressure-dependent regulation of Ca2+ signalling in the vascular endothelium. J Physiol 2015; 593:5231-53. [PMID: 26507455 PMCID: PMC4704526 DOI: 10.1113/jp271157] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2015] [Accepted: 10/19/2015] [Indexed: 12/19/2022] Open
Abstract
Key points Increased pressure suppresses endothelial control of vascular tone but it remains uncertain (1) how pressure is sensed by the endothelium and (2) how the vascular response is inhibited. This study used a novel imaging method to study large numbers of endothelial cells in arteries that were in a physiological configuration and held at normal blood pressures. Increased pressure suppressed endothelial IP3‐mediated Ca2+ signals. Pressure modulated endothelial cell shape. The changes in cell shape may alter endothelial Ca2+ signals by modulating the diffusive environment for Ca2+ near IP3 receptors. Endothelial pressure‐dependent mechanosensing may occur without a requirement for a conventional molecular mechanoreceptor.
Abstract The endothelium is an interconnected network upon which haemodynamic mechanical forces act to control vascular tone and remodelling in disease. Ca2+ signalling is central to the endothelium's mechanotransduction and networked activity. However, challenges in imaging Ca2+ in large numbers of endothelial cells under conditions that preserve the intact physical configuration of pressurized arteries have limited progress in understanding how pressure‐dependent mechanical forces alter networked Ca2+ signalling. We developed a miniature wide‐field, gradient‐index (GRIN) optical probe designed to fit inside an intact pressurized artery that permitted Ca2+ signals to be imaged with subcellular resolution in a large number (∼200) of naturally connected endothelial cells at various pressures. Chemical (acetylcholine) activation triggered spatiotemporally complex, propagating inositol trisphosphate (IP3)‐mediated Ca2+ waves that originated in clusters of cells and progressed from there across the endothelium. Mechanical stimulation of the artery, by increased intraluminal pressure, flattened the endothelial cells and suppressed IP3‐mediated Ca2+ signals in all activated cells. By computationally modelling Ca2+ release, endothelial shape changes were shown to alter the geometry of the Ca2+ diffusive environment near IP3 receptor microdomains to limit IP3‐mediated Ca2+ signals as pressure increased. Changes in cell shape produce a geometric microdomain regulation of IP3‐mediated Ca2+ signalling to explain macroscopic pressure‐dependent, endothelial mechanosensing without the need for a conventional mechanoreceptor. The suppression of IP3‐mediated Ca2+ signalling may explain the decrease in endothelial activity as pressure increases. GRIN imaging provides a convenient method that gives access to hundreds of endothelial cells in intact arteries in physiological configuration. Increased pressure suppresses endothelial control of vascular tone but it remains uncertain (1) how pressure is sensed by the endothelium and (2) how the vascular response is inhibited. This study used a novel imaging method to study large numbers of endothelial cells in arteries that were in a physiological configuration and held at normal blood pressures. Increased pressure suppressed endothelial IP3‐mediated Ca2+ signals. Pressure modulated endothelial cell shape. The changes in cell shape may alter endothelial Ca2+ signals by modulating the diffusive environment for Ca2+ near IP3 receptors. Endothelial pressure‐dependent mechanosensing may occur without a requirement for a conventional molecular mechanoreceptor.
Collapse
Affiliation(s)
- Calum Wilson
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, SIPBS Building, 161 Cathedral Street, Glasgow, G4 0RE, UK
| | - Christopher D Saunter
- Centre for Advanced Instrumentation, Biophysical Sciences Institute, Department of Physics, Durham University, South Road, Durham, DH1 3LE, UK
| | - John M Girkin
- Centre for Advanced Instrumentation, Biophysical Sciences Institute, Department of Physics, Durham University, South Road, Durham, DH1 3LE, UK
| | - John G McCarron
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, SIPBS Building, 161 Cathedral Street, Glasgow, G4 0RE, UK
| |
Collapse
|
43
|
Billcliff PG, Noakes CJ, Mehta ZB, Yan G, Mak L, Woscholski R, Lowe M. OCRL1 engages with the F-BAR protein pacsin 2 to promote biogenesis of membrane-trafficking intermediates. Mol Biol Cell 2015; 27:90-107. [PMID: 26510499 PMCID: PMC4694765 DOI: 10.1091/mbc.e15-06-0329] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2015] [Accepted: 10/23/2015] [Indexed: 12/26/2022] Open
Abstract
Mutation of the inositol 5-phosphatase OCRL1 causes Lowe syndrome and Dent-2 disease. Loss of OCRL1 function perturbs several cellular processes, including membrane traffic, but the underlying mechanisms remain poorly defined. Here we show that OCRL1 is part of the membrane-trafficking machinery operating at the trans-Golgi network (TGN)/endosome interface. OCRL1 interacts via IPIP27A with the F-BAR protein pacsin 2. OCRL1 and IPIP27A localize to mannose 6-phosphate receptor (MPR)-containing trafficking intermediates, and loss of either protein leads to defective MPR carrier biogenesis at the TGN and endosomes. OCRL1 5-phosphatase activity, which is membrane curvature sensitive, is stimulated by IPIP27A-mediated engagement of OCRL1 with pacsin 2 and promotes scission of MPR-containing carriers. Our data indicate a role for OCRL1, via IPIP27A, in regulating the formation of pacsin 2-dependent trafficking intermediates and reveal a mechanism for coupling PtdIns(4,5)P2 hydrolysis with carrier biogenesis on endomembranes.
Collapse
Affiliation(s)
- Peter G Billcliff
- Faculty of Life Sciences, University of Manchester, Manchester M13 9PT, United Kingdom
| | - Christopher J Noakes
- Faculty of Life Sciences, University of Manchester, Manchester M13 9PT, United Kingdom
| | - Zenobia B Mehta
- Faculty of Life Sciences, University of Manchester, Manchester M13 9PT, United Kingdom
| | - Guanhua Yan
- Faculty of Life Sciences, University of Manchester, Manchester M13 9PT, United Kingdom
| | - LokHang Mak
- Department of Chemistry, Imperial College, London SW7 2AZ, United Kingdom
| | - Rudiger Woscholski
- Department of Chemistry, Imperial College, London SW7 2AZ, United Kingdom
| | - Martin Lowe
- Faculty of Life Sciences, University of Manchester, Manchester M13 9PT, United Kingdom
| |
Collapse
|
44
|
Abstract
BAR proteins comprise a heterogeneous group of multi-domain proteins with diverse biological functions. The common denominator is the Bin-Amphiphysin-Rvs (BAR) domain that not only confers targeting to lipid bilayers, but also provides scaffolding to mold lipid membranes into concave or convex surfaces. This function of BAR proteins is an important determinant in the dynamic reconstruction of membrane vesicles, as well as of the plasma membrane. Several BAR proteins function as linkers between cytoskeletal regulation and membrane dynamics. These links are provided by direct interactions between BAR proteins and actin-nucleation-promoting factors of the Wiskott-Aldrich syndrome protein family and the Diaphanous-related formins. The Rho GTPases are key factors for orchestration of this intricate interplay. This review describes how BAR proteins regulate the activity of Rho GTPases, as well as how Rho GTPases regulate the function of BAR proteins. This mutual collaboration is a central factor in the regulation of vital cellular processes, such as cell migration, cytokinesis, intracellular transport, endocytosis, and exocytosis.
Collapse
Affiliation(s)
- Pontus Aspenström
- a Department of Microbiology and Tumor and Cell Biology; Karolinska Institutet ; Stockholm , Sweden
| |
Collapse
|
45
|
Stebbing J, Zhang H, Xu Y, Grothey A, Ajuh P, Angelopoulos N, Giamas G. Characterization of the Tyrosine Kinase-Regulated Proteome in Breast Cancer by Combined use of RNA interference (RNAi) and Stable Isotope Labeling with Amino Acids in Cell Culture (SILAC) Quantitative Proteomics. Mol Cell Proteomics 2015; 14:2479-92. [PMID: 26089344 PMCID: PMC4563730 DOI: 10.1074/mcp.m115.048090] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2015] [Indexed: 12/13/2022] Open
Abstract
Tyrosine kinases (TKs) are central regulators in cellular activities and perturbations of TK signaling contribute to oncogenesis. However, less than half of the TKs have been thoroughly studied and a global functional analysis of their proteomic portrait is lacking. Here we conducted a combined approach of RNA interference (RNAi) and stable isotope labeling with amino acids in cell culture (SILAC)-based quantitative proteomics to decode the TK-regulated proteome and associated signaling dynamics. As a result, a broad proteomic repertoire modulated by TKs was revealed, upon silencing of the 65 TKs expressed in MCF7 breast cancer cells. This yielded 10 new distinctive TK clusters according to similarity in TK-regulated proteome, each characterized by a unique signaling signature in contrast to previous classifications. We provide functional analyses and identify critical pathways for each cluster based on their common downstream targets. Analysis of different breast cancer subtypes showed distinct correlations of each cluster with clinical outcome. From the significantly up- and down-regulated proteins, we identified a number of markers of drug sensitivity and resistance. These data supports the role of TKs in regulating major aspects of cellular activity, but also reveals redundancy in signaling, explaining why kinase inhibitors alone often fail to achieve their clinical aims. The TK-SILACepedia provides a comprehensive resource for studying the global function of TKs in cancer.
Collapse
Affiliation(s)
- Justin Stebbing
- From the ‡Department of Surgery and Cancer, Division of Cancer, Imperial College London, Hammersmith Hospital Campus, Du Cane Road, London, W12 ONN, UK
| | - Hua Zhang
- From the ‡Department of Surgery and Cancer, Division of Cancer, Imperial College London, Hammersmith Hospital Campus, Du Cane Road, London, W12 ONN, UK;
| | - Yichen Xu
- From the ‡Department of Surgery and Cancer, Division of Cancer, Imperial College London, Hammersmith Hospital Campus, Du Cane Road, London, W12 ONN, UK
| | - Arnhild Grothey
- From the ‡Department of Surgery and Cancer, Division of Cancer, Imperial College London, Hammersmith Hospital Campus, Du Cane Road, London, W12 ONN, UK
| | - Paul Ajuh
- §Dundee Cell Products Ltd, James Lindsay Place, Dundee Technopole, Dundee, DD1 5JJ, UK
| | - Nicos Angelopoulos
- From the ‡Department of Surgery and Cancer, Division of Cancer, Imperial College London, Hammersmith Hospital Campus, Du Cane Road, London, W12 ONN, UK
| | - Georgios Giamas
- From the ‡Department of Surgery and Cancer, Division of Cancer, Imperial College London, Hammersmith Hospital Campus, Du Cane Road, London, W12 ONN, UK
| |
Collapse
|
46
|
Williamson RC, Cowell CAM, Reville T, Roper JA, Rendall TCS, Bass MD. Coronin-1C Protein and Caveolin Protein Provide Constitutive and Inducible Mechanisms of Rac1 Protein Trafficking. J Biol Chem 2015; 290:15437-15449. [PMID: 25925950 PMCID: PMC4505459 DOI: 10.1074/jbc.m115.640367] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2015] [Indexed: 02/05/2023] Open
Abstract
Sustained directional fibroblast migration requires both polarized activation of the protrusive signal, Rac1, and redistribution of inactive Rac1 from the rear of the cell so that it can be redistributed or degraded. In this work, we determine how alternative endocytic mechanisms dictate the fate of Rac1 in response to the extracellular matrix environment. We discover that both coronin-1C and caveolin retrieve Rac1 from similar locations at the rear and sides of the cell. We find that coronin-1C-mediated extraction, which is responsible for Rac1 recycling, is a constitutive process that maintains Rac1 protein levels within the cell. In the absence of coronin-1C, the effect of caveolin-mediated endocytosis, which targets Rac1 for proteasomal degradation, becomes apparent. Unlike constitutive coronin-1C-mediated trafficking, caveolin-mediated Rac1 endocytosis is induced by engagement of the fibronectin receptor syndecan-4. Such an inducible endocytic/degradation mechanism would predict that, in the presence of fibronectin, caveolin defines regions of the cell that are resistant to Rac1 activation but, in the absence of fibronectin leaves more of the membrane susceptible to Rac1 activation and protrusion. Indeed, we demonstrate that fibronectin-stimulated activation of Rac1 is accelerated in the absence of caveolin and that, when caveolin is knocked down, polarization of active Rac1 is lost in FRET experiments and culminates in shunting migration in a fibrous fibronectin matrix. Although the concept of polarized Rac1 activity in response to chemoattractants has always been apparent, our understanding of the balance between recycling and degradation explains how polarity can be maintained when the chemotactic gradient has faded.
Collapse
Affiliation(s)
- Rosalind C Williamson
- School of Biochemistry and University of Bristol, University Walk, Bristol BS8 1TD, United Kingdom
| | - Christopher A M Cowell
- School of Biochemistry and University of Bristol, University Walk, Bristol BS8 1TD, United Kingdom
| | - Thomas Reville
- School of Biochemistry and University of Bristol, University Walk, Bristol BS8 1TD, United Kingdom
| | - James A Roper
- School of Biochemistry and University of Bristol, University Walk, Bristol BS8 1TD, United Kingdom
| | - Thomas C S Rendall
- Department of Engineering, University of Bristol, University Walk, Bristol BS8 1TD, United Kingdom
| | - Mark D Bass
- School of Biochemistry and University of Bristol, University Walk, Bristol BS8 1TD, United Kingdom; Centre for Membrane Interactions and Dynamics, Department of Biomedical Science, University of Sheffield, Western Bank, Sheffield S10 2TN, United Kingdom.
| |
Collapse
|
47
|
Day CA, Baetz NW, Copeland CA, Kraft LJ, Han B, Tiwari A, Drake KR, De Luca H, Chinnapen DJF, Davidson MW, Holmes RK, Jobling MG, Schroer TA, Lencer WI, Kenworthy AK. Microtubule motors power plasma membrane tubulation in clathrin-independent endocytosis. Traffic 2015; 16:572-90. [PMID: 25690058 PMCID: PMC4440230 DOI: 10.1111/tra.12269] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2015] [Revised: 02/06/2015] [Accepted: 02/06/2015] [Indexed: 12/13/2022]
Abstract
How the plasma membrane is bent to accommodate clathrin-independent endocytosis remains uncertain. Recent studies suggest Shiga and cholera toxin induce membrane curvature required for their uptake into clathrin-independent carriers by binding and cross-linking multiple copies of their glycosphingolipid receptors on the plasma membrane. But it remains unclear if toxin-induced sphingolipid crosslinking provides sufficient mechanical force for deforming the plasma membrane, or if host cell factors also contribute to this process. To test this, we imaged the uptake of cholera toxin B-subunit into surface-derived tubular invaginations. We found that cholera toxin mutants that bind to only one glycosphingolipid receptor accumulated in tubules, and that toxin binding was entirely dispensable for membrane tubulations to form. Unexpectedly, the driving force for tubule extension was supplied by the combination of microtubules, dynein and dynactin, thus defining a novel mechanism for generating membrane curvature during clathrin-independent endocytosis.
Collapse
Affiliation(s)
- Charles A Day
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN, USA.,Current address: Hormel Institute, University of Minnesota, Austin, MN, USA
| | - Nicholas W Baetz
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Courtney A Copeland
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Lewis J Kraft
- Chemical and Physical Biology Program, Vanderbilt University, Nashville, TN, USA
| | - Bing Han
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Ajit Tiwari
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Kimberly R Drake
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Heidi De Luca
- GI Cell Biology, Department of Pediatrics, Boston Children's Hospital, Boston, MA, USA
| | - Daniel J-F Chinnapen
- GI Cell Biology, Department of Pediatrics, Boston Children's Hospital, Boston, MA, USA
| | - Michael W Davidson
- National High Magnetic Field Laboratory, The Florida State University, Tallahassee, FL, USA
| | - Randall K Holmes
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Michael G Jobling
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Trina A Schroer
- Department of Biology, The Johns Hopkins University, Baltimore, MD, USA
| | - Wayne I Lencer
- GI Cell Biology, Department of Pediatrics, Boston Children's Hospital, Boston, MA, USA.,Harvard Medical School and the Harvard Digestive Diseases Center, Boston, MA, USA
| | - Anne K Kenworthy
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN, USA.,Chemical and Physical Biology Program, Vanderbilt University, Nashville, TN, USA.,Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN, USA.,Epithelial Biology Program, Vanderbilt University School of Medicine, Nashville, TN, USA
| |
Collapse
|
48
|
FlnA binding to PACSIN2 F-BAR domain regulates membrane tubulation in megakaryocytes and platelets. Blood 2015; 126:80-8. [PMID: 25838348 DOI: 10.1182/blood-2014-07-587600] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2014] [Accepted: 03/30/2015] [Indexed: 11/20/2022] Open
Abstract
Bin-Amphiphysin-Rvs (BAR) and Fes-CIP4 homology BAR (F-BAR) proteins generate tubular membrane invaginations reminiscent of the megakaryocyte (MK) demarcation membrane system (DMS), which provides membranes necessary for future platelets. The F-BAR protein PACSIN2 is one of the most abundant BAR/F-BAR proteins in platelets and the only one reported to interact with the cytoskeletal and scaffold protein filamin A (FlnA), an essential regulator of platelet formation and function. The FlnA-PACSIN2 interaction was therefore investigated in MKs and platelets. PACSIN2 associated with FlnA in human platelets. The interaction required FlnA immunoglobulin-like repeat 20 and the tip of PACSIN2 F-BAR domain and enhanced PACSIN2 F-BAR domain membrane tubulation in vitro. Most human and wild-type mouse platelets had 1 to 2 distinct PACSIN2 foci associated with cell membrane GPIbα, whereas Flna-null platelets had 0 to 4 or more foci. Endogenous PACSIN2 and transfected enhanced green fluorescent protein-PACSIN2 were concentrated in midstage wild-type mouse MKs in a well-defined invagination of the plasma membrane reminiscent of the initiating DMS and dispersed in the absence of FlnA binding. The DMS appeared less well defined, and platelet territories were not readily visualized in Flna-null MKs. We conclude that the FlnA-PACSIN2 interaction regulates membrane tubulation in MKs and platelets and likely contributes to DMS formation.
Collapse
|
49
|
Havrylov S, Park M. MS/MS-based strategies for proteomic profiling of invasive cell structures. Proteomics 2014; 15:272-86. [PMID: 25303514 DOI: 10.1002/pmic.201400220] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2014] [Revised: 08/19/2014] [Accepted: 10/01/2014] [Indexed: 12/29/2022]
Abstract
Acquired capacity of cancer cells to penetrate through the extracellular matrix of surrounding tissues is a prerequisite for tumour metastatic spread - the main source of cancer-associated mortality. Through combined efforts of many research groups, we are beginning to understand that the ability of cells to invade through the extracellular matrix is a multi-faceted phenomenon supported by variety of specialised protrusive cellular structures, primarily pseudopodia, invadopodia and podosomes. Additionally, secreted extracellular vesicles are being increasingly recognised as important mediators of invasive cell phenotypes and therefore may be considered bona fide invasive cell structures. Dissection of the molecular makings underlying biogenesis and function of all of these structures is crucial to identify novel targets for specific anti-metastatic therapies. Rapid advances and growing accessibility of MS/MS-based protein identification made this family of techniques a suitable and appropriate choice for proteomic profiling of invasive cell structures. In this review, we provide a summary of current progress in the characterisation of protein composition and topology of protein interaction networks of pseudopodia, invadopodia, podosomes and extracellular vesicles, as well as outline challenges and perspectives of the field.
Collapse
Affiliation(s)
- Serhiy Havrylov
- Goodman Cancer Research Centre, McGill University, Montreal, QC, Canada; Department of Medicine, McGill University, Montreal, QC, Canada
| | | |
Collapse
|
50
|
Kostan J, Salzer U, Orlova A, Törö I, Hodnik V, Senju Y, Zou J, Schreiner C, Steiner J, Meriläinen J, Nikki M, Virtanen I, Carugo O, Rappsilber J, Lappalainen P, Lehto VP, Anderluh G, Egelman EH, Djinović-Carugo K. Direct interaction of actin filaments with F-BAR protein pacsin2. EMBO Rep 2014; 15:1154-62. [PMID: 25216944 DOI: 10.15252/embr.201439267] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Two mechanisms have emerged as major regulators of membrane shape: BAR domain-containing proteins, which induce invaginations and protrusions, and nuclear promoting factors, which cause generation of branched actin filaments that exert mechanical forces on membranes. While a large body of information exists on interactions of BAR proteins with membranes and regulatory proteins of the cytoskeleton, little is known about connections between these two processes. Here, we show that the F-BAR domain protein pacsin2 is able to associate with actin filaments using the same concave surface employed to bind to membranes, while some other tested N-BAR and F-BAR proteins (endophilin, CIP4 and FCHO2) do not associate with actin. This finding reveals a new level of complexity in membrane remodeling processes.
Collapse
Affiliation(s)
- Julius Kostan
- Department of Structural and Computational Biology, Max F. Perutz Laboratories, University of Vienna, Vienna, Austria
| | - Ulrich Salzer
- Department of Structural and Computational Biology, Max F. Perutz Laboratories, University of Vienna, Vienna, Austria Department of Medical Biochemistry, Medical University of Vienna, Vienna, Austria
| | - Albina Orlova
- Department of Biochemistry and Molecular Genetics, University of Virginia Medical Center, Charlottesville, VA, USA
| | - Imre Törö
- Department of Structural and Computational Biology, Max F. Perutz Laboratories, University of Vienna, Vienna, Austria
| | - Vesna Hodnik
- Department of Biology, Biotechnical Faculty, University of Ljubljana, Ljubljana, Slovenia
| | - Yosuke Senju
- Program in Cell and Molecular Biology, Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - Juan Zou
- Wellcome Trust Centre for Cell Biology, University of Edinburgh, Edinburgh, UK
| | - Claudia Schreiner
- Department of Structural and Computational Biology, Max F. Perutz Laboratories, University of Vienna, Vienna, Austria
| | - Julia Steiner
- Department of Structural and Computational Biology, Max F. Perutz Laboratories, University of Vienna, Vienna, Austria
| | - Jari Meriläinen
- Department of Pathology, Haartman Institute, University of Helsinki, Helsinki, Finland
| | - Marko Nikki
- Department of Pathology, Haartman Institute, University of Helsinki, Helsinki, Finland
| | - Ismo Virtanen
- Institute of Biomedicine/Anatomy, University of Helsinki, Helsinki, Finland
| | - Oliviero Carugo
- Department of Structural and Computational Biology, Max F. Perutz Laboratories, University of Vienna, Vienna, Austria Department of Chemistry, University of Pavia, Pavia, Italy
| | - Juri Rappsilber
- Department of Pathology, Haartman Institute, University of Helsinki, Helsinki, Finland Department of Biotechnology, Technological University of Berlin, Berlin, Germany
| | - Pekka Lappalainen
- Program in Cell and Molecular Biology, Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - Veli-Pekka Lehto
- Department of Pathology, Haartman Institute, University of Helsinki, Helsinki, Finland
| | - Gregor Anderluh
- Department of Biology, Biotechnical Faculty, University of Ljubljana, Ljubljana, Slovenia National Institute of Chemistry, Ljubljana, Slovenia EN-FIST Centre of Excellence, Ljubljana, Slovena
| | - Edward H Egelman
- Department of Biochemistry and Molecular Genetics, University of Virginia Medical Center, Charlottesville, VA, USA
| | - Kristina Djinović-Carugo
- Department of Structural and Computational Biology, Max F. Perutz Laboratories, University of Vienna, Vienna, Austria Department of Biochemistry, Faculty of Chemistry and Chemical Technology, University of Ljubljana, Ljubljana, Slovenia
| |
Collapse
|