1
|
Stephens CA, van Hilten N, Zheng L, Grabe M. Simulation-based survey of TMEM16 family reveals that robust lipid scrambling requires an open groove. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.09.25.615027. [PMID: 39386458 PMCID: PMC11463437 DOI: 10.1101/2024.09.25.615027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/12/2024]
Abstract
Biological membranes are complex and dynamic structures with different populations of lipids in their inner and outer leaflets. The Ca2+-activated TMEM16 family of membrane proteins plays an important role in collapsing this asymmetric lipid distribution by spontaneously, and bidirectionally, scrambling phospholipids between the two leaflets, which can initiate signaling and alter the physical properties of the membrane. While evidence shows that lipid scrambling can occur via an open hydrophilic pathway ("groove") that spans the membrane, it remains unclear if all family members facilitate lipid movement in this manner. Here we present a comprehensive computational study of lipid scrambling by all TMEM16 members with experimentally solved structures. We performed coarse-grained molecular dynamics (MD) simulations of 27 structures from five different family members solved under activating and non-activating conditions, and we captured over 700 scrambling events in aggregate. This enabled us to directly compare scrambling rates, mechanisms, and protein-lipid interactions for fungal and mammalian TMEM16s, in both open (Ca2+-bound) and closed (Ca2+-free) conformations with statistical rigor. We show that all TMEM16 structures thin the membrane and that the majority of scrambling (>90%) occurs at the groove only when TM4 and TM6 have sufficiently separated. Surprisingly, we also observed 60 scrambling events that occurred outside the canonical groove, over 90% of which took place at the dimer-dimer interface in mammalian TMEM16s. This new site suggests an alternative mechanism for lipid scrambling in the absence of an open groove.
Collapse
Affiliation(s)
- Christina A. Stephens
- Cardiovascular Research Institute, University of California, San Francisco, CA 94158
- Graduate Group in Biophysics, University of California, San Francisco, CA 94158
- Department of Pharmaceutical Chemistry, University of California, San Francisco, CA 94158
| | - Niek van Hilten
- Cardiovascular Research Institute, University of California, San Francisco, CA 94158
- Department of Pharmaceutical Chemistry, University of California, San Francisco, CA 94158
| | - Lisa Zheng
- Cardiovascular Research Institute, University of California, San Francisco, CA 94158
- Graduate Group in Biophysics, University of California, San Francisco, CA 94158
- Department of Pharmaceutical Chemistry, University of California, San Francisco, CA 94158
| | - Michael Grabe
- Cardiovascular Research Institute, University of California, San Francisco, CA 94158
| |
Collapse
|
2
|
Lian Z, Luo W, Liu J, Wang J, Chai W, Wang Y, Sethi S, Ma X. Analysis of ANO6, HAPLN1, and EDIL3 Polymorphisms in Patients with Ankylosing Spondylitis in a Chinese Han Population: A Case-Control Study. Genet Test Mol Biomarkers 2024; 28:385-392. [PMID: 39358671 DOI: 10.1089/gtmb.2023.0569] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/04/2024] Open
Abstract
Background: Earlier research has demonstrated a genetic basis for the susceptibility to ankylosing spondylitis (AS) and the severity of AS. By employing a genome-wide association study, recent work has established a correlation between the susceptibility to AS and the ANO6, HAPLN, and EDIL3 genes in a Western study population-though alternative studies have not corroborated these findings. This study aims to examine the effects of ANO6, HAPLN1, and EDIL3 polymorphisms on the susceptibility and severity of AS among the predominantly Chinese Han population. Methods: The study involved the collection of blood samples from 497 patients with AS and 498 nonrelated healthy individuals. All participants in the study were human leukocyte antigen (HLA) HLA-B27 positive and of Han Chinese descent. Illness severity was the criteria used for classifying patients with AS. Thirteen tagSNPs in ANO6, HAPLN1, and EDIL3 were chosen and then subjected to genetic typing. Analysis was conducted on the occurrence rates of various genotypes and alleles between the control group and patients with varying AS severity. Results: Following Bonferroni correction, it was found that the rs4768085 and rs17095830 single nucleotide polymorphism (SNPs) in ANO6 were related to the susceptibility to AS. Further, the rs6869296 SNP in HAPLN1 and the rs2301071 SNP between EDIL3 and HAPLN1 were also related to AS susceptibility. Regarding AS severity, the rs4768085, rs2897868, rs7965430, and rs11182965 SNPs in ANO6 were found to be associated. Conclusions: Among the Han population in China, the ANO6 and HAPLN1 genes are related to the susceptibility to AS; the ANO6 gene is also associated with the severity of AS.
Collapse
Affiliation(s)
- Zijian Lian
- Department of Orthopaedics, Tianjin Hospital, Tianjin, China
| | - Wei Luo
- Department of Orthopaedics, Tianjin Hospital, Tianjin, China
| | - Jun Liu
- Department of Orthopaedics, Tianjin Hospital, Tianjin, China
| | - Jing Wang
- Department of Orthopaedics, Tianjin Hospital, Tianjin, China
| | - Wei Chai
- Department of Orthopaedics, Chinese People's Liberation Army General Hospital, Beijing, China
| | - Yan Wang
- Department of Orthopaedics, Chinese People's Liberation Army General Hospital, Beijing, China
| | - Sahil Sethi
- Department of Orthopaedics, University of Chicago Hospital, Chicago, Illinois, USA
| | - Xinlong Ma
- Department of Orthopaedics, Tianjin Hospital, Tianjin, China
| |
Collapse
|
3
|
Schreiber R, Ousingsawat J, Kunzelmann K. The anoctamins: Structure and function. Cell Calcium 2024; 120:102885. [PMID: 38642428 DOI: 10.1016/j.ceca.2024.102885] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Revised: 04/03/2024] [Accepted: 04/04/2024] [Indexed: 04/22/2024]
Abstract
When activated by increase in intracellular Ca2+, anoctamins (TMEM16 proteins) operate as phospholipid scramblases and as ion channels. Anoctamin 1 (ANO1) is the Ca2+-activated epithelial anion-selective channel that is coexpressed together with the abundant scramblase ANO6 and additional intracellular anoctamins. In salivary and pancreatic glands, ANO1 is tightly packed in the apical membrane and secretes Cl-. Epithelia of airways and gut use cystic fibrosis transmembrane conductance regulator (CFTR) as an apical Cl- exit pathway while ANO1 supports Cl- secretion mainly by facilitating activation of luminal CFTR and basolateral K+ channels. Under healthy conditions ANO1 modulates intracellular Ca2+ signals by tethering the endoplasmic reticulum, and except of glands its direct secretory contribution as Cl- channel might be small, compared to CFTR. In the kidneys ANO1 supports proximal tubular acid secretion and protein reabsorption and probably helps to excrete HCO3-in the collecting duct epithelium. However, under pathological conditions as in polycystic kidney disease, ANO1 is strongly upregulated and may cause enhanced proliferation and cyst growth. Under pathological condition, ANO1 and ANO6 are upregulated and operate as secretory channel/phospholipid scramblases, partly by supporting Ca2+-dependent processes. Much less is known about the role of other epithelial anoctamins whose potential functions are discussed in this review.
Collapse
Affiliation(s)
- Rainer Schreiber
- Physiological Institute, University of Regensburg, University street 31, D-93053 Regensburg, Germany
| | - Jiraporn Ousingsawat
- Physiological Institute, University of Regensburg, University street 31, D-93053 Regensburg, Germany
| | - Karl Kunzelmann
- Physiological Institute, University of Regensburg, University street 31, D-93053 Regensburg, Germany.
| |
Collapse
|
4
|
Prouse T, Majumder R. TMEM16E: unscrambling our knowledge about coagulation. J Thromb Haemost 2023; 21:3000-3004. [PMID: 37633641 DOI: 10.1016/j.jtha.2023.08.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 08/14/2023] [Accepted: 08/16/2023] [Indexed: 08/28/2023]
Affiliation(s)
- Teagan Prouse
- Louisiana State University Health Sciences Center, New Orleans, LA, USA
| | - Rinku Majumder
- Louisiana State University Health Sciences Center, New Orleans, LA, USA.
| |
Collapse
|
5
|
Li X, Zhong Z, Zhang R, Zhang J, Zhang Y, Zeng S, Du Q, Wang H, Zhang S, Lu L, Li M, Long K. Decoding the transcriptome of muscular dystrophy due to Ptrf deficiency using single-nucleus RNA sequencing. FASEB J 2023; 37:e22993. [PMID: 37235502 DOI: 10.1096/fj.202201949rr] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 04/20/2023] [Accepted: 05/12/2023] [Indexed: 05/28/2023]
Abstract
Lacking PTRF (polymerase I and transcript release factor), an essential caveolae component, causes a secondary deficiency of caveolins resulting in muscular dystrophy. The transcriptome responses of different types of muscle fibers and mononuclear cells in skeletal muscle to muscular dystrophy caused by Ptrf deletion have not been explored. Here, we created muscular dystrophy mice by Ptrf knockout and applied single-nucleus RNA sequencing (snRNA-seq) to unveil the transcriptional changes of the skeletal muscle at single-nucleus resolution. 11 613 muscle nuclei (WT, 5838; Ptrf KO, 5775) were classified into 12 clusters corresponding to 11 nuclear types. Trajectory analysis revealed the potential transition between type IIb_1 and IIb_2 myonuclei upon muscular dystrophy. Functional enrichment analysis indicated that apoptotic signaling and enzyme-linked receptor protein signaling pathway were significantly enriched in type IIb_1 and IIb_2 myonuclei of Ptrf KO, respectively. The muscle structure development and the PI3K-AKT signaling pathway were significantly enriched in type IIa and IIx myonuclei of Ptrf KO. Meanwhile, metabolic pathway analysis showed a decrease in overall metabolic pathway activity of myonuclei subtypes upon muscular dystrophy, with the most decrease in type IIb_1 myonuclei. Gene regulatory network analysis found that the activity of Mef2c, Mef2d, Myf5, and Pax3 regulons was enhanced in type II myonuclei of Ptrf KO, especially in type IIb_2 myonuclei. In addition, we investigated the transcriptome changes in adipocytes and found that muscular dystrophy enhanced the lipid metabolic capacity of adipocytes. Our findings provide a valuable resource for exploring the molecular mechanism of muscular dystrophy due to Ptrf deficiency.
Collapse
Affiliation(s)
- Xiaokai Li
- Livestock and Poultry Multi-omics Key Laboratory of Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China
| | - Zhining Zhong
- Livestock and Poultry Multi-omics Key Laboratory of Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China
| | - Ruowei Zhang
- Livestock and Poultry Multi-omics Key Laboratory of Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China
| | - Jiaman Zhang
- Livestock and Poultry Multi-omics Key Laboratory of Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China
| | - Yu Zhang
- Livestock and Poultry Multi-omics Key Laboratory of Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China
| | - Sha Zeng
- Livestock and Poultry Multi-omics Key Laboratory of Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China
| | - Qinjiao Du
- Livestock and Poultry Multi-omics Key Laboratory of Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China
| | - Haoming Wang
- Livestock and Poultry Multi-omics Key Laboratory of Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China
| | - Songling Zhang
- Livestock and Poultry Multi-omics Key Laboratory of Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China
| | - Lu Lu
- Livestock and Poultry Multi-omics Key Laboratory of Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China
| | - Mingzhou Li
- Livestock and Poultry Multi-omics Key Laboratory of Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China
| | - Keren Long
- Livestock and Poultry Multi-omics Key Laboratory of Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China
| |
Collapse
|
6
|
Schmaier AA, Anderson PF, Chen SM, El-Darzi E, Aivasovsky I, Kaushik MP, Sack KD, Hartzell HC, Parikh SM, Flaumenhaft R, Schulman S. TMEM16E regulates endothelial cell procoagulant activity and thrombosis. J Clin Invest 2023; 133:e163808. [PMID: 36951953 PMCID: PMC10231993 DOI: 10.1172/jci163808] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Accepted: 03/22/2023] [Indexed: 03/24/2023] Open
Abstract
Endothelial cells (ECs) normally form an anticoagulant surface under physiological conditions, but switch to support coagulation following pathogenic stimuli. This switch promotes thrombotic cardiovascular disease. To generate thrombin at physiologic rates, coagulation proteins assemble on a membrane containing anionic phospholipid, most notably phosphatidylserine (PS). PS can be rapidly externalized to the outer cell membrane leaflet by phospholipid "scramblases," such as TMEM16F. TMEM16F-dependent PS externalization is well characterized in platelets. In contrast, how ECs externalize phospholipids to support coagulation is not understood. We employed a focused genetic screen to evaluate the contribution of transmembrane phospholipid transport on EC procoagulant activity. We identified 2 TMEM16 family members, TMEM16F and its closest paralog, TMEM16E, which were both required to support coagulation on ECs via PS externalization. Applying an intravital laser-injury model of thrombosis, we observed, unexpectedly, that PS externalization was concentrated at the vessel wall, not on platelets. TMEM16E-null mice demonstrated reduced vessel-wall-dependent fibrin formation. The TMEM16 inhibitor benzbromarone prevented PS externalization and EC procoagulant activity and protected mice from thrombosis without increasing bleeding following tail transection. These findings indicate the activated endothelial surface is a source of procoagulant phospholipid contributing to thrombus formation. TMEM16 phospholipid scramblases may be a therapeutic target for thrombotic cardiovascular disease.
Collapse
Affiliation(s)
- Alec A. Schmaier
- Division of Cardiovascular Medicine and
- Division of Hemostasis and Thrombosis, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, USA
| | | | | | - Emale El-Darzi
- Division of Hemostasis and Thrombosis, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, USA
| | | | | | - Kelsey D. Sack
- Division of Pulmonary, Critical Care and Sleep Medicine, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
| | - H. Criss Hartzell
- Department of Cell Biology, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Samir M. Parikh
- Division of Nephrology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, USA
- Division of Nephrology and Departments of Internal Medicine and Pharmacology, University of Texas Southwestern Medical School, Dallas, Texas, USA
| | - Robert Flaumenhaft
- Division of Hemostasis and Thrombosis, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, USA
- Division of Hematology and Hematologic Malignancies, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, USA
| | - Sol Schulman
- Division of Hemostasis and Thrombosis, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, USA
- Division of Hematology and Hematologic Malignancies, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
7
|
Boekell KL, Brown BJ, Talbot BE, Schlondorff JS. Trpc6 gain-of-function disease mutation enhances phosphatidylserine exposure in murine platelets. PLoS One 2022; 17:e0270431. [PMID: 35749414 PMCID: PMC9231752 DOI: 10.1371/journal.pone.0270431] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Accepted: 06/09/2022] [Indexed: 01/28/2023] Open
Abstract
Platelets enhance coagulation by exposing phosphatidylserine (PS) on their cell surface in response to strong agonist activation. Transient receptor potential channels, including TRPC6, have been implicated in the calcium influx central to this process. Here, we characterize the effect of a Trpc6 gain-of-function (GOF) disease-associated, and a dominant negative (DN), mutation on murine platelet activation. Platelets from mice harboring Trpc6E896K/E896K (GOF) and Trpc6DN/DN mutations were subject to in vitro analysis. Trpc6E896K/E896K and Trpc6DN/DN mutant platelets show enhanced and absent calcium influx, respectively, upon addition of the TRPC3/6 agonist GSK1702934A (GSK). GSK was sufficient to induce integrin αIIbβ3 activation, P-selection and PS exposure, talin cleavage, and MLC2 phosphorylation in Trpc6E896K/E896K, but not in wild-type, platelets. Thrombin-induced calcium influx and PS exposure were enhanced, and clot retraction delayed, by GOF TRPC6, while no differences were noted between wild-type and Trpc6DN/DN platelets. In contrast, Erk activation upon GSK treatment was absent in Trpc6DN/DN, and enhanced in Trpc6E896K/E896K, platelets, compared to wild-type. The positive allosteric modulator, TRPC6-PAM-C20, and fluoxetine maintained their ability to enhance and inhibit, respectively, GSK-mediated calcium influx in Trpc6E896K/E896K platelets. The data demonstrate that gain-of-function mutant TRPC6 channel can enhance platelet activation, including PS exposure, while confirming that TRPC6 is not necessary for this process. Furthermore, the results suggest that Trpc6 GOF disease mutants do not simply increase wild-type TRPC6 responses, but can affect pathways not usually modulated by TRPC6 channel activity, displaying a true gain-of-function phenotype.
Collapse
Affiliation(s)
- Kimber L. Boekell
- Department of Medicine, Division of Nephrology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Brittney J. Brown
- Department of Medicine, Division of Nephrology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Brianna E. Talbot
- Department of Medicine, Division of Nephrology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Johannes S. Schlondorff
- Department of Medicine, Division of Nephrology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, United States of America
| |
Collapse
|
8
|
Hu W, Li M, Zhang Q, Liu C, Wang X, Li J, Qiu S, Li L. Establishment of a novel CNV-related prognostic signature predicting prognosis in patients with breast cancer. J Ovarian Res 2021; 14:103. [PMID: 34364397 PMCID: PMC8349487 DOI: 10.1186/s13048-021-00823-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Accepted: 05/10/2021] [Indexed: 01/17/2023] Open
Abstract
BACKGROUND Copy number variation (CNVs) is a key factor in breast cancer development. This study determined prognostic molecular characteristics to predict breast cancer through performing a comprehensive analysis of copy number and gene expression data. METHODS Breast cancer expression profiles, CNV and complete information from The Cancer Genome Atlas (TCGA) dataset were collected. Gene Expression Omnibus (GEO) chip data sets (GSE20685 and GSE31448) containing breast cancer samples were used as external validation sets. Univariate survival COX analysis, multivariate survival COX analysis, least absolute shrinkage and selection operator (LASSO), Chi square, Kaplan-Meier (KM) survival curve and receiver operating characteristic (ROC) analysis were applied to build a gene signature model and assess its performance. RESULTS A total of 649 CNV related-differentially expressed gene obtained from TCGA-breast cancer dataset were related to several cancer pathways and functions. A prognostic gene sets with 9 genes were developed to stratify patients into high-risk and low-risk groups, and its prognostic performance was verified in two independent patient cohorts (n = 327, 246). The result uncovered that 9-gene signature could independently predict breast cancer prognosis. Lower mutation of PIK3CA and higher mutation of TP53 and CDH1 were found in samples with high-risk score compared with samples with low-risk score. Patients in the high-risk group showed higher immune score, malignant clinical features than those in the low-risk group. The 9-gene signature developed in this study achieved a higher AUC. CONCLUSION The current research established a 5-CNV gene signature to evaluate prognosis of breast cancer patients, which may innovate clinical application of prognostic assessment.
Collapse
Affiliation(s)
- Wei Hu
- Department of Thyroid and Breast Surgery, Zibo Central Hospital, Zibo, 255036, China
| | - Mingyue Li
- Department of Rehabilitation Medicine, The Third Affilated Hospital, Sun Yat-sen University, Guangzhou, 510000, China
| | - Qi Zhang
- Blood Transfusion Department, Zibo Central Hospital, Zibo, 255036, China
| | - Chuan Liu
- Department of Thyroid and Breast Surgery, Zibo Central Hospital, Zibo, 255036, China
| | - Xinmei Wang
- Department of Pathology, ZiBo Central Hospital, Zibo, 255036, China
| | - Jing Li
- Department of Pathology, ZiBo Central Hospital, Zibo, 255036, China.
| | - Shusheng Qiu
- Department of Thyroid and Breast Surgery, Zibo Central Hospital, Zibo, 255036, China.
| | - Liang Li
- Department of Thyroid and Breast Surgery, Zibo Central Hospital, Zibo, 255036, China.
| |
Collapse
|
9
|
Proteomic Studies of Primary Acute Myeloid Leukemia Cells Derived from Patients Before and during Disease-Stabilizing Treatment Based on All-Trans Retinoic Acid and Valproic Acid. Cancers (Basel) 2021; 13:cancers13092143. [PMID: 33946813 PMCID: PMC8125016 DOI: 10.3390/cancers13092143] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 04/16/2021] [Accepted: 04/20/2021] [Indexed: 12/18/2022] Open
Abstract
All-trans retinoic acid (ATRA) and valproic acid (VP) have been tried in the treatment of non-promyelocytic variants of acute myeloid leukemia (AML). Non-randomized studies suggest that the two drugs can stabilize AML and improve normal peripheral blood cell counts. In this context, we used a proteomic/phosphoproteomic strategy to investigate the in vivo effects of ATRA/VP on human AML cells. Before starting the combined treatment, AML responders showed increased levels of several proteins, especially those involved in neutrophil degranulation/differentiation, M phase regulation and the interconversion of nucleotide di- and triphosphates (i.e., DNA synthesis and binding). Several among the differentially regulated phosphorylation sites reflected differences in the regulation of RNA metabolism and apoptotic events at the same time point. These effects were mainly caused by increased cyclin dependent kinase 1 and 2 (CDK1/2), LIM domain kinase 1 and 2 (LIMK1/2), mitogen-activated protein kinase 7 (MAPK7) and protein kinase C delta (PRKCD) activity in responder cells. An extensive effect of in vivo treatment with ATRA/VP was the altered level and phosphorylation of proteins involved in the regulation of transcription/translation/RNA metabolism, especially in non-responders, but the regulation of cell metabolism, immune system and cytoskeletal functions were also affected. Our analysis of serial samples during the first week of treatment suggest that proteomic and phosphoproteomic profiling can be used for the early identification of responders to ATRA/VP-based treatment.
Collapse
|
10
|
Öhlinger T, Müllner EW, Fritz M, Sauer T, Werning M, Baron DM, Salzer U. Lysophosphatidic acid-induced pro-thrombotic phosphatidylserine exposure and ionophore-induced microvesiculation is mediated by the scramblase TMEM16F in erythrocytes. Blood Cells Mol Dis 2020; 83:102426. [PMID: 32222693 DOI: 10.1016/j.bcmd.2020.102426] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Revised: 03/12/2020] [Accepted: 03/12/2020] [Indexed: 12/31/2022]
Abstract
Recent studies indicate that erythrocytes actively modulate blood clotting and thrombus formation. The lipid mediator lysophosphatidic acid (LPA) is produced by activated platelets, and triggers a signaling process in erythrocytes. This results in cellular calcium uptake and exposure of phosphatidylserine (PS) at the cell surface, thereby generating activated membrane binding sites for factors of the clotting cascade. Moreover, erythrocytes of patients with a bleeding disorder and mutations in the scramblase TMEM16F show impaired PS exposure and microvesiculation upon treatment with calcium ionophore. We report that TMEM16F inhibitors tannic acid (TA) and epigallocatechin-3-gallate (EGCG) inhibit LPA-induced PS exposure and calcium uptake at low micromolar concentrations; fluoxetine, an antidepressant and a known activator of TMEM16F, enhances these processes. These effectors likewise modulate erythrocyte PS exposure and microvesicle shedding induced by calcium ionophore treatment. Further, LPA-treated erythrocytes triggered thrombin generation in platelet-free plasma which was partially impaired in the presence of TA and EGCG. Thus, this study suggests that LPA activates the scramblase TMEM16F in erythrocytes, thereby possibly mediating a pro-thrombotic function in these cells. EGCG as well as fluoxetine, substances with potentially high plasma concentrations due to alimentation or medical treatment, should be considered as potential effectors of systemic hemostatic regulation.
Collapse
Affiliation(s)
- Thomas Öhlinger
- Center for Medical Biochemistry, Max Perutz Laboratories, Medical University of Vienna, Vienna, Austria; Department of Anaesthesia, Intensive Care Medicine and Pain Medicine, Medical University of Vienna, Vienna, Austria
| | - Ernst W Müllner
- Center for Medical Biochemistry, Max Perutz Laboratories, Medical University of Vienna, Vienna, Austria
| | - Magdalena Fritz
- Center for Medical Biochemistry, Max Perutz Laboratories, Medical University of Vienna, Vienna, Austria
| | - Thomas Sauer
- Center for Medical Biochemistry, Max Perutz Laboratories, Medical University of Vienna, Vienna, Austria
| | - Maike Werning
- Center for Medical Biochemistry, Max Perutz Laboratories, Medical University of Vienna, Vienna, Austria
| | - David M Baron
- Department of Anaesthesia, Intensive Care Medicine and Pain Medicine, Medical University of Vienna, Vienna, Austria
| | - Ulrich Salzer
- Center for Medical Biochemistry, Max Perutz Laboratories, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
11
|
Kunisaki C. Role of the Anoctamin Family in Various Carcinomas. Ann Surg Oncol 2020; 27:3112-3114. [PMID: 32347400 DOI: 10.1245/s10434-020-08371-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2020] [Indexed: 12/17/2022]
Affiliation(s)
- Chikara Kunisaki
- Department of Surgery, Gastroenterological Center, Yokohama City University, Yokohama, Japan.
| |
Collapse
|
12
|
Xuan ZB, Wang YJ, Xie J. ANO6 promotes cell proliferation and invasion in glioma through regulating the ERK signaling pathway. Onco Targets Ther 2019; 12:6721-6731. [PMID: 31692479 PMCID: PMC6708391 DOI: 10.2147/ott.s211725] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Accepted: 06/11/2019] [Indexed: 12/19/2022] Open
Abstract
Purpose Anoctamin6 (ANO6) plays a crucial role in several cancers, whereas the specific role of ANO6 in glioblastoma is unclear. Methods Kaplan-Meier survival analysis was used to analysis the correlation between ANO6 and survival rate of patients with glioblastoma. Univariate Cox regression analysis was used to analysis the correlation among ANO6 expression level,and age, gender, WHO and overall survival rate. Immunohistocemical technique, RT-PCR and western blot were used to dected the ANO6 expression. CCK8, colony formation and transwell were used to detected cell viability, cell proliferation and cell invasion in glioblastoma cells transfected with sh-ANO6 and ANO6 overexpression. In addition, after SHG-44 cells trasfected with ANO6 overexpression were ERK inhibitor (PD98059), CCK8, colony formation and transwell were used to detected cell viability, cell proliferation and cell invasion. Western blot was used to detected ERK protein level and the phosphorylation level of ERK in T89G and U87MG cells tranfected wih sh-ANO6. Results The results indicated that the ANO6 expression level was significantly associated with patients' age and tumor stage. Univariate Cox regression analysis showed that the ANO6 expression level, age, gender and tumor stage were not related to the overall survival rate. ANO6 inhibition significantly suppressed the viability, invasion and the ability of colony formation in glioma cells, while ANO6 overexpression led to the opposite results in SHG-44 cells. ANO6 knockdown strongly inhibits the phosphorylation level and nuclear translocation of extracellular signal-regulated kinase (ERK) protein to inhibit ERK signaling. ERK inhibitor significantly decreased the cell proliferation and invasion in SHG-44 cells transfected with sh-ANO6. Conclusion This study revealed that ANO6 activited ERK signaling pathway through promoting the nuclear translocation of ERK to increase the proliferation and invasion of glioblastoma cells.
Collapse
Affiliation(s)
- Zhao-Bo Xuan
- Department of Neurosurgery, The First Hospital Affiliated to Jiamusi University, Jiamusi City, Heilongjiang Province 154002, People's Republic of China
| | - Ye-Ji Wang
- Department of Neurosurgery, Shanxian Haijiya Hospital, Heze City, Shandong Province 274300, People's Republic of China
| | - Jun Xie
- Department of Neurosurgery, Tongchuan People's Hospital, Tongchuan City, Shaanxi Province 727000, People's Republic of China
| |
Collapse
|
13
|
Benedetto R, Ousingsawat J, Cabrita I, Pinto M, Lérias JR, Wanitchakool P, Schreiber R, Kunzelmann K. Plasma membrane-localized TMEM16 proteins are indispensable for expression of CFTR. J Mol Med (Berl) 2019; 97:711-722. [PMID: 30915480 DOI: 10.1007/s00109-019-01770-4] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Revised: 02/22/2019] [Accepted: 03/05/2019] [Indexed: 10/27/2022]
Abstract
The cystic fibrosis transmembrane conductance regulator (CFTR) is the secretory chloride channel in epithelial tissues that has a central role in cystic fibrosis (CF) lung and gastrointestinal disease. A recent publication demonstrates a close association between CFTR and TMEM16A, the calcium-activated chloride channel. Thus, no CFTR chloride currents could be detected in airways and large intestine from mice lacking epithelial expression of TMEM16A. Here, we demonstrate that another plasma membrane-localized TMEM16 paralogue, TMEM16F, can compensate for the lack of TMEM16A. Using TMEM16 knockout mice, human lymphocytes, and a number of human cell lines with endogenous protein expression or heterologous expression, we demonstrate that CFTR can only function in the presence of either TMEM16A or TMEM16F. Double knockout of intestinal epithelial TMEM16A/F expression did not produce offsprings, suggesting a lethal phenotype in utero. Plasma membrane-localized TMEM16A or TMEM16F is required for exocytosis and expression of CFTR in the plasma membrane. TMEM16A/F proteins may therefore have an impact on disease severity in CF. KEY MESSAGES: • Cystic fibrosis is caused by the defective Cl- channel cystic fibrosis transmembrane conductance regulator (CFTR). • A close relationship exists between CFTR and the calcium-activated chloride channels TMEM16A/TMEM16F. • In conditional airway and intestinal knockout mice, lymphocytes from Scott disease patients and in overexpressing cells, CFTR is not functional in the absence of TMEM16A and TMEM16F. • TMEM16A and TMEM16F support membrane exocytosis and are essential for plasma membrane insertion of CFTR.
Collapse
Affiliation(s)
- Roberta Benedetto
- Physiological Institute, University of Regensburg, University Street 31, D-93053, Regensburg, Germany
| | - Jiraporn Ousingsawat
- Physiological Institute, University of Regensburg, University Street 31, D-93053, Regensburg, Germany
| | - Inês Cabrita
- Physiological Institute, University of Regensburg, University Street 31, D-93053, Regensburg, Germany
| | - Madalena Pinto
- Physiological Institute, University of Regensburg, University Street 31, D-93053, Regensburg, Germany
| | - Joana R Lérias
- Physiological Institute, University of Regensburg, University Street 31, D-93053, Regensburg, Germany
| | - Podchanart Wanitchakool
- Physiological Institute, University of Regensburg, University Street 31, D-93053, Regensburg, Germany
| | - Rainer Schreiber
- Physiological Institute, University of Regensburg, University Street 31, D-93053, Regensburg, Germany
| | - Karl Kunzelmann
- Physiological Institute, University of Regensburg, University Street 31, D-93053, Regensburg, Germany.
| |
Collapse
|
14
|
Lin H, Jun I, Woo JH, Lee MG, Kim SJ, Nam JH. Temperature-dependent increase in the calcium sensitivity and acceleration of activation of ANO6 chloride channel variants. Sci Rep 2019; 9:6706. [PMID: 31040335 PMCID: PMC6491614 DOI: 10.1038/s41598-019-43162-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Accepted: 04/12/2019] [Indexed: 02/08/2023] Open
Abstract
Anoctamin-6 (ANO6) belongs to a family of calcium (Ca2+)-activated chloride channels (CaCCs), with three splicing variants (V1, V2, and V5) showing plasma membrane expression. Unlike other CaCCs, ANO6 requires a non-physiological intracellular free calcium concentration ([Ca2+]i > 1 μM) and several minutes for full activation under a whole-cell patch clamp. Therefore, its physiological role as an ion channel is uncertain and it is more commonly considered a Ca2+-dependent phospholipid scramblase. Here, we demonstrate that physiological temperature (37 °C) increases ANO6 Ca2+ sensitivity under a whole-cell patch clamp; V1 was activated by 1 μM [Ca2+]i, whereas V2 and V5 were activated by 300 nM [Ca2+]i. Increasing the temperature to 42 °C led to activation of all ANO6 variants by 100 nM [Ca2+]i. The delay time for activation of the three variants was significantly shortened at 37 °C. Notably, the temperature-dependent Ca2+-sensitisation of ANO6 became insignificant under inside-out patch clamp, suggesting critical roles of unknown cytosolic factors. Unlike channel activity, 27 °C but not 37 °C (physiological temperature) induced the scramblase activity of ANO6 at submicromolar [Ca2+]i (300 nM), irrespective of variant type. Our results reveal a physiological ion conducting property of ANO6 at 37 °C and suggest that ANO6 channel function acts separately from its scramblase activity.
Collapse
Affiliation(s)
- Haiyue Lin
- Department of Physiology, Seoul National University College of Medicine, 103 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea
| | - Ikhyun Jun
- The Institute of Vision Research, Department of Ophthalmology, Yonsei University College of Medicine, 50 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
- Department of Pharmacology and Brain Korea 21 PLUS Project for Medical Sciences, Yonsei University College of Medicine, 50 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Joo Han Woo
- Department of Physiology, Dongguk University College of Medicine, 123 Dongdae-ro, Gyeongju, 38066, Republic of Korea
- Channelopathy Research Center (CRC), Dongguk University College of Medicine, 32 Dongguk-ro, Ilsan Dong-gu, Goyang, Gyeonggi-do, 10326, Republic of Korea
| | - Min Goo Lee
- Department of Pharmacology and Brain Korea 21 PLUS Project for Medical Sciences, Yonsei University College of Medicine, 50 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Sung Joon Kim
- Department of Physiology, Seoul National University College of Medicine, 103 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea.
| | - Joo Hyun Nam
- Department of Physiology, Dongguk University College of Medicine, 123 Dongdae-ro, Gyeongju, 38066, Republic of Korea.
- Channelopathy Research Center (CRC), Dongguk University College of Medicine, 32 Dongguk-ro, Ilsan Dong-gu, Goyang, Gyeonggi-do, 10326, Republic of Korea.
| |
Collapse
|
15
|
Le T, Le SC, Yang H. Drosophila Subdued is a moonlighting transmembrane protein 16 (TMEM16) that transports ions and phospholipids. J Biol Chem 2019; 294:4529-4537. [PMID: 30700552 DOI: 10.1074/jbc.ac118.006530] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Revised: 01/25/2019] [Indexed: 12/19/2022] Open
Abstract
Transmembrane protein 16 (TMEM16) family members play numerous important physiological roles, ranging from controlling membrane excitability and secretion to mediating blood coagulation and viral infection. These diverse functions are largely due to their distinct biophysical properties. Mammalian TMEM16A and TMEM16B are Ca2+-activated Cl- channels (CaCCs), whereas mammalian TMEM16F, fungal afTMEM16, and nhTMEM16 are moonlighting (multifunctional) proteins with both Ca2+-activated phospholipid scramblase (CaPLSase) and Ca2+-activated, nonselective ion channel (CAN) activities. To further understand the biological functions of the enigmatic TMEM16 proteins in different organisms, here, by combining an improved annexin V-based CaPLSase-imaging assay with inside-out patch clamp technique, we thoroughly characterized Subdued, a Drosophila TMEM16 ortholog. We show that Subdued is also a moonlighting transport protein with both CAN and CaPLSase activities. Using a TMEM16F-deficient HEK293T cell line to avoid strong interference from endogenous CaPLSases, our functional characterization and mutagenesis studies revealed that Subdued is a bona fide CaPLSase. Our finding that Subdued is a moonlighting TMEM16 expands our understanding of the molecular mechanisms of TMEM16 proteins and their evolution and physiology in both Drosophila and humans.
Collapse
Affiliation(s)
- Trieu Le
- From the Departments of Biochemistry and
| | - Son C Le
- From the Departments of Biochemistry and
| | - Huanghe Yang
- From the Departments of Biochemistry and .,Neurobiology, Duke University Medical Center, Durham, North Carolina 27710
| |
Collapse
|
16
|
Janks L, Sprague RS, Egan TM. ATP-Gated P2X7 Receptors Require Chloride Channels To Promote Inflammation in Human Macrophages. THE JOURNAL OF IMMUNOLOGY 2018; 202:883-898. [PMID: 30598517 DOI: 10.4049/jimmunol.1801101] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Accepted: 11/25/2018] [Indexed: 12/28/2022]
Abstract
Immune cells of myeloid origin show robust expression of ATP-gated P2X7 receptors, two-transmembrane ion channels permeable to Na+, K+, and Ca2+ Receptor activation promotes inflammasome activation and release of the proinflammatory cytokines IL-1β and IL-18. In this study, we show that ATP generates facilitating cationic currents in monocyte-derived human macrophages and permeabilizes the plasma membrane to polyatomic cationic dyes. We find that antagonists of PLA2 and Cl- channels abolish P2X7 receptor-mediated current facilitation, membrane permeabilization, blebbing, phospholipid scrambling, inflammasome activation, and IL-1β release. Our data demonstrate significant differences in the actions of ATP in murine and human macrophages and suggest that PLA2 and Cl- channels mediate innate immunity downstream of P2X7 receptors in human macrophages.
Collapse
Affiliation(s)
- Laura Janks
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine, St. Louis, MO 63104
| | - Randy S Sprague
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine, St. Louis, MO 63104
| | - Terrance M Egan
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine, St. Louis, MO 63104
| |
Collapse
|
17
|
Abstract
Mucociliary clearance is critically important in protecting the airways from infection and from the harmful effects of smoke and various inspired substances known to induce oxidative stress and persistent inflammation. An essential feature of the clearance mechanism involves regulation of the periciliary liquid layer on the surface of the airway epithelium, which is necessary for normal ciliary beating and maintenance of mucus hydration. The underlying ion transport processes associated with airway surface hydration include epithelial Na+ channel-dependent Na+ absorption occurring in parallel with CFTR and Ca2+-activated Cl- channel-dependent anion secretion, which are coordinately regulated to control the depth of the periciliary liquid layer. Oxidative stress is known to cause both acute and chronic effects on airway ion transport function, and an increasing number of studies in the past few years have identified an important role for autophagy as part of the physiological response to the damaging effects of oxidation. In this review, recent studies addressing the influence of oxidative stress and autophagy on airway ion transport pathways, along with results showing the potential of autophagy modulators in restoring the function of ion channels involved in transepithelial electrolyte transport necessary for effective mucociliary clearance, are presented.
Collapse
Affiliation(s)
- Scott M O'Grady
- Departments of Animal Science, Integrative Biology and Physiology, University of Minnesota , St. Paul, Minnesota
| |
Collapse
|
18
|
Schenk LK, Ousingsawat J, Skryabin BV, Schreiber R, Pavenstädt H, Kunzelmann K. Regulation and Function of TMEM16F in Renal Podocytes. Int J Mol Sci 2018; 19:ijms19061798. [PMID: 29912162 PMCID: PMC6032267 DOI: 10.3390/ijms19061798] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2018] [Revised: 06/12/2018] [Accepted: 06/14/2018] [Indexed: 12/13/2022] Open
Abstract
The Ca2+-activated phospholipid scramblase and ion channel TMEM16F is expressed in podocytes of renal glomeruli. Podocytes are specialized cells that form interdigitating foot processes as an essential component of the glomerular filter. These cells, which participate in generation of the primary urine, are often affected during primary glomerular diseases, such as glomerulonephritis and secondary hypertensive or diabetic nephropathy, which always leads to proteinuria. Because the function of podocytes is known to be controlled by intracellular Ca2+ signaling, it is important to know about the role of Ca2+-activated TMEM16F in these cells. To that end, we generated an inducible TMEM16F knockdown in the podocyte cell line AB8, and produced a conditional mouse model with knockout of TMEM16F in podocytes and renal epithelial cells of the nephron. We found that knockdown of TMEM16F did not produce proteinuria or any obvious phenotypic changes. Knockdown of TMEM16F affected cell death of tubular epithelial cells but not of glomerular podocytes when analyzed in TUNEL assays. Surprisingly, and in contrast to other cell types, TMEM16F did not control intracellular Ca2+ signaling and was not responsible for Ca2+-activated whole cell currents in podocytes. TMEM16F levels in podocytes were enhanced after inhibition of the endolysosomal pathway and after treatment with angiotensin II. Renal knockout of TMEM16F did not compromise renal morphology and serum electrolytes. Taken together, in contrast to other cell types, such as platelets, bone cells, and immune cells, TMEM16F shows little effect on basal properties of podocytes and does not appear to be essential for renal function.
Collapse
Affiliation(s)
- Laura K Schenk
- Department of Nephrology, Hypertension and Rheumatology, University Hospital Muenster, 48149 Muenster, Germany.
| | - Jiraporn Ousingsawat
- Institut für Physiologie, Universität Regensburg, Universitätsstraße 31, D-93053 Regensburg, Germany.
| | - Boris V Skryabin
- Transgenic Animal and Genetic Engineering Models (TRAM), Department of Medicine, Westfälischen, Wilhelms⁻Universität Münster, 48149 Münster, Germany.
| | - Rainer Schreiber
- Institut für Physiologie, Universität Regensburg, Universitätsstraße 31, D-93053 Regensburg, Germany.
| | - Hermann Pavenstädt
- Department of Nephrology, Hypertension and Rheumatology, University Hospital Muenster, 48149 Muenster, Germany.
| | - Karl Kunzelmann
- Institut für Physiologie, Universität Regensburg, Universitätsstraße 31, D-93053 Regensburg, Germany.
| |
Collapse
|
19
|
Falzone ME, Malvezzi M, Lee BC, Accardi A. Known structures and unknown mechanisms of TMEM16 scramblases and channels. J Gen Physiol 2018; 150:933-947. [PMID: 29915161 PMCID: PMC6028493 DOI: 10.1085/jgp.201711957] [Citation(s) in RCA: 81] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2018] [Accepted: 05/29/2018] [Indexed: 12/25/2022] Open
Abstract
Falzone et al. interpret the mechanisms underlying the activity of TMEM16 family members from recent structural and functional work. The TMEM16 family of membrane proteins is composed of both Ca2+-gated Cl− channels and Ca2+-dependent phospholipid scramblases. The functional diversity of TMEM16s underlies their involvement in numerous signal transduction pathways that connect changes in cytosolic Ca2+ levels to cellular signaling networks. Indeed, defects in the function of several TMEM16s cause a variety of genetic disorders, highlighting their fundamental pathophysiological importance. Here, we review how our mechanistic understanding of TMEM16 function has been shaped by recent functional and structural work. Remarkably, the recent determination of near-atomic-resolution structures of TMEM16 proteins of both functional persuasions has revealed how relatively minimal rearrangements in the substrate translocation pathway are sufficient to precipitate the dramatic functional differences that characterize the family. These structures, when interpreted in the light of extensive functional analysis, point to an unusual mechanism for Ca2+-dependent activation of TMEM16 proteins in which substrate permeation is regulated by a combination of conformational rearrangements and electrostatics. These breakthroughs pave the way to elucidate the mechanistic bases of ion and lipid transport by the TMEM16 proteins and unravel the molecular links between these transport activities and their function in human pathophysiology.
Collapse
Affiliation(s)
- Maria E Falzone
- Department of Biochemistry, Weill Cornell Medical School, New York, NY
| | - Mattia Malvezzi
- Department of Anesthesiology, Weill Cornell Medical School, New York, NY
| | - Byoung-Cheol Lee
- Department of Anesthesiology, Weill Cornell Medical School, New York, NY
| | - Alessio Accardi
- Department of Biochemistry, Weill Cornell Medical School, New York, NY .,Department of Anesthesiology, Weill Cornell Medical School, New York, NY.,Department of Physiology, Biophysics and Systems Biology, Weill Cornell Medical School, New York, NY
| |
Collapse
|
20
|
Contribution of TMEM16F to pyroptotic cell death. Cell Death Dis 2018; 9:300. [PMID: 29463790 PMCID: PMC5833444 DOI: 10.1038/s41419-018-0373-8] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2017] [Revised: 02/02/2018] [Accepted: 02/05/2018] [Indexed: 02/08/2023]
Abstract
Pyroptosis is a highly inflammatory form of programmed cell death that is caused by infection with intracellular pathogens and activation of canonical or noncanonical inflammasomes. The purinergic receptor P2X7 is activated by the noncanonical inflammasome and contributes essentially to pyroptotic cell death. The Ca2+ activated phospholipid scramblase and ion channel TMEM16F has been shown earlier to control cellular effects downstream of purinergic P2X7 receptors that ultimately lead to cell death. As pyroptotic cell death is accompanied by an increases in intracellular Ca2+, we asked whether TMEM16F is activated during pyroptosis. The N-terminal cleavage product of gasdermin D (GD-N) is an executioner of pyroptosis by forming large plasma membrane pores. Expression of GD-N enhanced basal Ca2+ levels and induced cell death. We observed that GD-N induced cell death in HEK293 and HAP1 cells, which was depending on expression of endogenous TMEM16F. GD-N activated large whole cell currents that were suppressed by knockdown or inhibition of TMEM16F. The results suggest that whole cell currents induced by the pore forming domain of gasdermin-D, are at least in part due to activation of TMEM16F. Knockdown of other TMEM16 paralogues expressed in HAP1 cells suggest TMEM16F as a crucial element during pyroptosis and excluded a role of other TMEM16 proteins. Thus TMEM16F supports pyroptosis and other forms of inflammatory cell death such as ferroptosis. Its potent inhibition by tannic acid may be part of the anti-inflammatory effects of flavonoids.
Collapse
|
21
|
Schreiber R, Ousingsawat J, Wanitchakool P, Sirianant L, Benedetto R, Reiss K, Kunzelmann K. Regulation of TMEM16A/ANO1 and TMEM16F/ANO6 ion currents and phospholipid scrambling by Ca 2+ and plasma membrane lipid. J Physiol 2018; 596:217-229. [PMID: 29134661 PMCID: PMC5767690 DOI: 10.1113/jp275175] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Accepted: 11/08/2017] [Indexed: 12/12/2022] Open
Abstract
KEY POINTS TMEM16 proteins can operate as Ca2+ -activated Cl- channels or scramble membrane phospholipids, which are both highly relevant mechanisms during disease. Overexpression of TMEM16A and TMEM16F were found to be partially active at 37°C and at resting intracellular Ca2+ concentrations. We show that TMEM16 Cl- currents and phospholipid scrambling can be activated by modification of plasma membrane phospholipids, through reactive oxygen species and phospholipase A2. While phospholipids and Cl- ions are likely to use the same pore within TMEM16F, TMEM16A only conducts Cl- ions. Lipid regulation of TMEM16 proteins is highly relevant during inflammation and regulated cell death such as apoptosis and ferroptosis. ABSTRACT TMEM16/anoctamin (ANO) proteins form Ca2+ -activated ion channels or phospholipid scramblases. We found that both TMEM16A/ANO1 and TMEM16F/ANO6 produced Cl- currents when activated by intracellular Ca2+ , but only TMEM16F was able to expose phosphatidylserine to the outer leaflet of the plasma membrane. Mutations within TMEM16F or TMEM16A/F chimeras similarly changed Cl- currents and phospholipid scrambling, suggesting the same intramolecular pathway for Cl- and phospholipids. When overexpressed, TMEM16A and TMEM16F produced spontaneous Cl- currents at 37°C even at resting intracellular Ca2+ levels, which was abolished by inhibition of phospholipase A2 (PLA2 ). Connversely, activation of PLA2 or application of active PLA2 , as well as lipid peroxidation induced by reactive oxygen species (ROS) using staurosporine or tert-butyl hydroperoxide, enhanced ion currents by TMEM16A/F and in addition activated phospholipid scrambling by TMEM16F. Thus, TMEM16 proteins are activated by an increase in intracellular Ca2+ , or independent of intracellular Ca2+ , by modifications occurring in plasma and intracellular membrane phospholipids. These results may help to explain why regions distant to the TMEM16 pore and the Ca2+ binding sites control Cl- currents and phospholipid scrambling. Regulation of TMEM16 proteins through modification of membrane phospholipids occurs during regulated cell death such as apoptosis and ferroptosis. It contributes to inflammatory and nerve injury-induced hypersensitivity and generation of pain and therefore provides a regulatory mechanism that is particularly relevant during disease.
Collapse
Affiliation(s)
- Rainer Schreiber
- Institut für PhysiologieUniversität RegensburgUniversitätsstraße 31D‐93053RegensburgGermany
| | - Jiraporn Ousingsawat
- Institut für PhysiologieUniversität RegensburgUniversitätsstraße 31D‐93053RegensburgGermany
| | | | - Lalida Sirianant
- Institut für PhysiologieUniversität RegensburgUniversitätsstraße 31D‐93053RegensburgGermany
| | - Roberta Benedetto
- Institut für PhysiologieUniversität RegensburgUniversitätsstraße 31D‐93053RegensburgGermany
| | - Karina Reiss
- Department of DermatologyUniversity of KielSchittenhelmstrasse 7Kiel24105Germany
| | - Karl Kunzelmann
- Institut für PhysiologieUniversität RegensburgUniversitätsstraße 31D‐93053RegensburgGermany
| |
Collapse
|
22
|
Endosomal Trafficking Defects Can Induce Calcium-Dependent Azole Tolerance in Candida albicans. Antimicrob Agents Chemother 2016; 60:7170-7177. [PMID: 27645241 DOI: 10.1128/aac.01034-16] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2016] [Accepted: 09/11/2016] [Indexed: 01/16/2023] Open
Abstract
The azole antifungals arrest fungal growth through inhibition of ergosterol biosynthesis. We recently reported that a Candida albicans vps21Δ/Δ mutant, deficient in membrane trafficking through the late endosome/prevacuolar compartment (PVC), continues to grow in the presence of the azoles despite the depletion of cellular ergosterol. Here, we report that the vps21Δ/Δ mutant exhibits less plasma membrane damage upon azole treatment than the wild type, as measured by the release of a cytoplasmic luciferase reporter into the culture supernatant. Our results also reveal that the vps21Δ/Δ mutant has abnormal levels of intracellular Ca2+ and, in the presence of fluconazole, enhanced expression of a calcineurin-responsive RTA2-GFP reporter. Furthermore, the azole tolerance phenotype of the vps21Δ/Δ mutant is dependent upon both extracellular calcium levels and calcineurin activity. These findings underscore the importance of endosomal trafficking in determining the cellular consequences of azole treatment and indicate that this may occur through modulation of calcium- and calcineurin-dependent responses.
Collapse
|
23
|
Whitlock JM, Hartzell HC. Anoctamins/TMEM16 Proteins: Chloride Channels Flirting with Lipids and Extracellular Vesicles. Annu Rev Physiol 2016; 79:119-143. [PMID: 27860832 DOI: 10.1146/annurev-physiol-022516-034031] [Citation(s) in RCA: 133] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Anoctamin (ANO)/TMEM16 proteins exhibit diverse functions in cells throughout the body and are implicated in several human diseases. Although the founding members ANO1 (TMEM16A) and ANO2 (TMEM16B) are Ca2+-activated Cl- channels, most ANO paralogs are Ca2+-dependent phospholipid scramblases that serve as channels facilitating the movement (scrambling) of phospholipids between leaflets of the membrane bilayer. Phospholipid scrambling significantly alters the physical properties of the membrane and its landscape and has vast downstream signaling consequences. In particular, phosphatidylserine exposed on the external leaflet of the plasma membrane functions as a ligand for receptors vital for cell-cell communication. A major consequence of Ca2+-dependent scrambling is the release of extracellular vesicles that function as intercellular messengers by delivering signaling proteins and noncoding RNAs to alter target cell function. We discuss the physiological implications of Ca2+-dependent phospholipid scrambling, the extracellular vesicles associated with this activity, and the roles of ANOs in these processes.
Collapse
Affiliation(s)
- Jarred M Whitlock
- Department of Cell Biology, Emory University School of Medicine, Atlanta, Georgia 30322;
| | - H Criss Hartzell
- Department of Cell Biology, Emory University School of Medicine, Atlanta, Georgia 30322;
| |
Collapse
|
24
|
Gajek A, Denel-Bobrowska M, Rogalska A, Bukowska B, Maszewski J, Marczak A. Early Activation of Apoptosis and Caspase-independent Cell Death Plays an Important Role in Mediating the Cytotoxic and Genotoxic Effects of WP 631 in Ovarian Cancer Cells. Asian Pac J Cancer Prev 2016; 16:8503-12. [PMID: 26745109 DOI: 10.7314/apjcp.2015.16.18.8503] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
The purpose of this study was to provide a detailed explanation of the mechanism of bisanthracycline,?WP 631 in comparison to doxorubicin (DOX), a first generation anthracycline, currently the most widely used pharmaceutical in clinical oncology. Experiments were performed in SKOV-3 ovarian cancer cells which are otherwise resistant to standard drugs such as cis-platinum and adriamycin. As attention was focused on the ability of WP 631 to induce apoptosis, this was examined using a double staining method with Annexin V and propidium iodide probes, with measurement of the level of intracellular calcium ions and cytosolic cytochrome c. The western blotting technique was performed to confirm PARP cleavage. We also investigated the involvement of caspase activation and DNA degradation (comet assay and immunocytochemical detection of phosphorylated H2AX histones) in the development of apoptotic events. WP 631 demonstrated significantly higher effectiveness as a pro-apoptotic drug than DOX. This was evident in the higher levels of markers of apoptosis, such as the externalization of phosphatidylserine and the elevated level of cytochrome c. An extension of incubation time led to an increase in intracellular calcium levels after treatment with DOX. Lower changes in the calcium content were associated with the influence of WP 631. DOX led to the activation of all tested caspases, 8, 9 and 3, whereas WP 631 only induced an increase in caspase 8 activity after 24h of treatment and consequently led to the cleavage of PARP. The lack of active caspase 3 had no outcome on the single and double-stranded DNA breaks. The obtained results show that WP 631 was considerably more genotoxic towards the investigated cell line than DOX. This effect was especially visible after longer times of incubation. The above detailed studies indicate that WP 631 generates early apoptosis and cell death independent of caspase-3, detected at relatively late time points. The observed differences in the mechanisms of the action of WP631 and DOX suggest that this bisanthracycline can be an effective alternative in ovarian cancer treatment.
Collapse
Affiliation(s)
- Arkadiusz Gajek
- Department of Thermobiology, Faculty of Biology and Environmental Protection, University of Lodz, Pomorska, Lodz, Poland E-mail :
| | | | | | | | | | | |
Collapse
|
25
|
Wanitchakool P, Ousingsawat J, Sirianant L, MacAulay N, Schreiber R, Kunzelmann K. Cl - channels in apoptosis. EUROPEAN BIOPHYSICS JOURNAL : EBJ 2016; 45:599-610. [PMID: 27270446 DOI: 10.1007/s00249-016-1140-3] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/19/2016] [Revised: 05/06/2016] [Accepted: 05/14/2016] [Indexed: 12/28/2022]
Abstract
A remarkable feature of apoptosis is the initial massive cell shrinkage, which requires opening of ion channels to allow release of K+, Cl-, and organic osmolytes to drive osmotic water movement and cell shrinkage. This article focuses on the role of the Cl- channels LRRC8, TMEM16/anoctamin, and cystic fibrosis transmembrane conductance regulator (CFTR) in cellular apoptosis. LRRC8A-E has been identified as a volume-regulated anion channel expressed in many cell types. It was shown to be required for regulatory and apoptotic volume decrease (RVD, AVD) in cultured cell lines. Its presence also determines sensitivity towards cytostatic drugs such as cisplatin. Recent data point to a molecular and functional relationship of LRRC8A and anoctamins (ANOs). ANO6, 9, and 10 (TMEM16F, J, and K) augment apoptotic Cl- currents and AVD, but it remains unclear whether these anoctamins operate as Cl- channels or as regulators of other apoptotic Cl- channels, such as LRRC8. CFTR has been known for its proapoptotic effects for some time, and this effect may be based on glutathione release from the cell and increase in cytosolic reactive oxygen species (ROS). Although we find that CFTR is activated by cell swelling, it is possible that CFTR serves RVD/AVD through accumulation of ROS and activation of independent membrane channels such as ANO6. Thus activation of ANO6 will support cell shrinkage and induce additional apoptotic events, such as membrane phospholipid scrambling.
Collapse
Affiliation(s)
- Podchanart Wanitchakool
- Institut für Physiologie, Universität Regensburg, Universitätsstraße 31, 93053, Regensburg, Germany
| | - Jiraporn Ousingsawat
- Institut für Physiologie, Universität Regensburg, Universitätsstraße 31, 93053, Regensburg, Germany
| | - Lalida Sirianant
- Institut für Physiologie, Universität Regensburg, Universitätsstraße 31, 93053, Regensburg, Germany
| | - Nanna MacAulay
- Department of Cellular and Molecular Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Rainer Schreiber
- Institut für Physiologie, Universität Regensburg, Universitätsstraße 31, 93053, Regensburg, Germany
| | - Karl Kunzelmann
- Institut für Physiologie, Universität Regensburg, Universitätsstraße 31, 93053, Regensburg, Germany.
| |
Collapse
|
26
|
Solari FA, Mattheij NJA, Burkhart JM, Swieringa F, Collins PW, Cosemans JMEM, Sickmann A, Heemskerk JWM, Zahedi RP. Combined Quantification of the Global Proteome, Phosphoproteome, and Proteolytic Cleavage to Characterize Altered Platelet Functions in the Human Scott Syndrome. Mol Cell Proteomics 2016; 15:3154-3169. [PMID: 27535140 PMCID: PMC5054341 DOI: 10.1074/mcp.m116.060368] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2016] [Indexed: 11/19/2022] Open
Abstract
The Scott syndrome is a very rare and likely underdiagnosed bleeding disorder associated with mutations in the gene encoding anoctamin-6. Platelets from Scott patients are impaired in various Ca2+-dependent responses, including phosphatidylserine exposure, integrin closure, intracellular protein cleavage, and cytoskeleton-dependent morphological changes. Given the central role of anoctamin-6 in the platelet procoagulant response, we used quantitative proteomics to understand the underlying molecular mechanisms and the complex phenotypic changes in Scott platelets compared with control platelets. Therefore, we applied an iTRAQ-based multi-pronged strategy to quantify changes in (1) the global proteome, (2) the phosphoproteome, and (3) proteolytic events between resting and stimulated Scott and control platelets. Our data indicate a limited number of proteins with decreased (70) or increased (64) expression in Scott platelets, among those we confirmed the absence of anoctamin-6 and the strong up-regulation of aquaporin-1 by parallel reaction monitoring. The quantification of 1566 phosphopeptides revealed major differences between Scott and control platelets after stimulation with thrombin/convulxin or ionomycin. In Scott platelets, phosphorylation levels of proteins regulating cytoskeletal or signaling events were increased. Finally, we quantified 1596 N-terminal peptides in activated Scott and control platelets, 180 of which we identified as calpain-regulated, whereas a distinct set of 23 neo-N termini was caspase-regulated. In Scott platelets, calpain-induced cleavage of cytoskeleton-linked and signaling proteins was downregulated, in accordance with an increased phosphorylation state. Thus, multipronged proteomic profiling of Scott platelets provides detailed insight into their protection against detrimental Ca2+-dependent changes that are normally associated with phosphatidylserine exposure.
Collapse
Affiliation(s)
- Fiorella A Solari
- From the ‡Leibniz-Institut für Analytische Wissenschaften-ISAS-e.V., Dortmund, Germany
| | - Nadine J A Mattheij
- §Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands
| | - Julia M Burkhart
- From the ‡Leibniz-Institut für Analytische Wissenschaften-ISAS-e.V., Dortmund, Germany
| | - Frauke Swieringa
- §Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands
| | - Peter W Collins
- ¶Arthur Bloom Haemophilia Centre, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - Judith M E M Cosemans
- §Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands
| | - Albert Sickmann
- From the ‡Leibniz-Institut für Analytische Wissenschaften-ISAS-e.V., Dortmund, Germany; ‖Medizinisches Proteom-Center, Ruhr-University Bochum, Bochum, Germany; **Department of Chemistry, College of Physical Sciences, University of Aberdeen, Aberdeen, UK
| | - Johan W M Heemskerk
- §Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands;
| | - René P Zahedi
- From the ‡Leibniz-Institut für Analytische Wissenschaften-ISAS-e.V., Dortmund, Germany;
| |
Collapse
|
27
|
Abstract
Activation of ion channels and pores are essential steps during regulated cell death. Channels and pores participate in execution of apoptosis, necroptosis and other forms of caspase-independent cell death. Within the program of regulated cell death, these channels are strategically located. Ion channels can shrink cells and drive them towards apoptosis, resulting in silent, i.e. immunologically unrecognized cell death. Alternatively, activation of channels can induce cell swelling, disintegration of the cell membrane, and highly immunogenic necrotic cell death. The underlying cell death pathways are not strictly separated as identical stimuli may induce cell shrinkage and apoptosis when applied at low strength, but may also cause cell swelling at pronounced stimulation, resulting in regulated necrosis. Nevertheless, the precise role of ion channels during regulated cell death is far from being understood, as identical channels may support regulated death in some cell types, but may cause cell proliferation, cancer development, and metastasis in others. Along this line, the phospholipid scramblase and Cl(-)/nonselective channel anoctamin 6 (ANO6) shows interesting features, as it participates in apoptotic cell death during lower levels of activation, thereby inducing cell shrinkage. At strong activation, e.g. by stimulation of purinergic P2Y7 receptors, it participates in pore formation, causes massive membrane blebbing, cell swelling, and membrane disintegration. The LRRC8 proteins deserve much attention as they were found to have a major role in volume regulation, apoptotic cell shrinkage and resistance towards anticancer drugs.
Collapse
Affiliation(s)
- Karl Kunzelmann
- Institut für Physiologie, Universität Regensburg, Universitätsstraße 31, 93053, Regensburg, Germany.
| |
Collapse
|
28
|
van Geffen JP, Swieringa F, Heemskerk JW. Platelets and coagulation in thrombus formation: aberrations in the Scott syndrome. Thromb Res 2016; 141 Suppl 2:S12-6. [DOI: 10.1016/s0049-3848(16)30355-3] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|
29
|
Bevers EM, Williamson PL. Getting to the Outer Leaflet: Physiology of Phosphatidylserine Exposure at the Plasma Membrane. Physiol Rev 2016; 96:605-45. [PMID: 26936867 DOI: 10.1152/physrev.00020.2015] [Citation(s) in RCA: 330] [Impact Index Per Article: 36.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Phosphatidylserine (PS) is a major component of membrane bilayers whose change in distribution between inner and outer leaflets is an important physiological signal. Normally, members of the type IV P-type ATPases spend metabolic energy to create an asymmetric distribution of phospholipids between the two leaflets, with PS confined to the cytoplasmic membrane leaflet. On occasion, membrane enzymes, known as scramblases, are activated to facilitate transbilayer migration of lipids, including PS. Recently, two proteins required for such randomization have been identified: TMEM16F, a scramblase regulated by elevated intracellular Ca(2+), and XKR8, a caspase-sensitive protein required for PS exposure in apoptotic cells. Once exposed at the cell surface, PS regulates biochemical reactions involved in blood coagulation, and bone mineralization, and also regulates a variety of cell-cell interactions. Exposed on the surface of apoptotic cells, PS controls their recognition and engulfment by other cells. This process is exploited by parasites to invade their host, and in specialized form is used to maintain photoreceptors in the eye and modify synaptic connections in the brain. This review discusses what is known about the mechanism of PS exposure at the surface of the plasma membrane of cells, how actors in the extracellular milieu sense surface exposed PS, and how this recognition is translated to downstream consequences of PS exposure.
Collapse
Affiliation(s)
- Edouard M Bevers
- Department of Biochemistry, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, The Netherlands; and Department of Biology, Amherst College, Amherst, Massachusetts
| | - Patrick L Williamson
- Department of Biochemistry, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, The Netherlands; and Department of Biology, Amherst College, Amherst, Massachusetts
| |
Collapse
|
30
|
Mattheij NJA, Braun A, van Kruchten R, Castoldi E, Pircher J, Baaten CCFMJ, Wülling M, Kuijpers MJE, Köhler R, Poole AW, Schreiber R, Vortkamp A, Collins PW, Nieswandt B, Kunzelmann K, Cosemans JMEM, Heemskerk JWM. Survival protein anoctamin-6 controls multiple platelet responses including phospholipid scrambling, swelling, and protein cleavage. FASEB J 2016; 30:727-37. [PMID: 26481309 DOI: 10.1096/fj.15-280446] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2015] [Accepted: 10/05/2015] [Indexed: 11/11/2022]
Abstract
Scott syndrome is a rare bleeding disorder, characterized by altered Ca(2+)-dependent platelet signaling with defective phosphatidylserine (PS) exposure and microparticle formation, and is linked to mutations in the ANO6 gene, encoding anoctamin (Ano)6. We investigated how the complex platelet phenotype of this syndrome is linked to defective expression of Anos or other ion channels. Mice were generated with heterozygous of homozygous deficiency in Ano6, Ano1, or Ca(2+)-dependent KCa3.1 Gardos channel. Platelets from these mice were extensively analyzed on molecular functions and compared with platelets from a patient with Scott syndrome. Deficiency in Ano1 or Gardos channel did not reduce platelet responses compared with control mice (P > 0.1). In 2 mouse strains, deficiency in Ano6 resulted in reduced viability with increased bleeding time to 28.6 min (control 6.4 min, P < 0.05). Platelets from the surviving Ano6-deficient mice resembled platelets from patients with Scott syndrome in: 1) normal collagen-induced aggregate formation (P > 0.05) with reduced PS exposure (-65 to 90%); 2) lowered Ca(2+)-dependent swelling (-80%) and membrane blebbing (-90%); 3) reduced calpain-dependent protein cleavage (-60%); and 4) moderately affected apoptosis-dependent PS exposure. In conclusion, mouse deficiency of Ano6 but not of other channels affects viability and phenocopies the complex changes in platelets from hemostatically impaired patients with Scott syndrome.
Collapse
Affiliation(s)
- Nadine J A Mattheij
- *Department of Cell Biochemistry of Thrombosis and Haemostasis Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), University of Maastricht, Maastricht, The Netherlands; Department of Experimental Biomedicine, University Hospital and Rudolf Virchow Center, University of Würzburg, Würzburg, Germany; Walter Brendel Centre of Experimental Medicine and German Centre of Cardiovascular Research, Munich Heart Alliance, Ludwig-Maximilians-Universität München, München, Germany; Department of Developmental Biology, Centre for Medical Biotechnology, University of Duisburg-Essen, Duisburg-Essen, Germany; Aragon Institute of Health Sciences I+CS/IIS and ARAID, Zaragoza, Spain; School of Physiology and Pharmacology, University of Bristol, Bristol, United Kingdom; Institute of Physiology, University of Regensburg, Regensburg, Germany; **Arthur Bloom Haemophilia Centre, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - Attila Braun
- *Department of Cell Biochemistry of Thrombosis and Haemostasis Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), University of Maastricht, Maastricht, The Netherlands; Department of Experimental Biomedicine, University Hospital and Rudolf Virchow Center, University of Würzburg, Würzburg, Germany; Walter Brendel Centre of Experimental Medicine and German Centre of Cardiovascular Research, Munich Heart Alliance, Ludwig-Maximilians-Universität München, München, Germany; Department of Developmental Biology, Centre for Medical Biotechnology, University of Duisburg-Essen, Duisburg-Essen, Germany; Aragon Institute of Health Sciences I+CS/IIS and ARAID, Zaragoza, Spain; School of Physiology and Pharmacology, University of Bristol, Bristol, United Kingdom; Institute of Physiology, University of Regensburg, Regensburg, Germany; **Arthur Bloom Haemophilia Centre, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - Roger van Kruchten
- *Department of Cell Biochemistry of Thrombosis and Haemostasis Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), University of Maastricht, Maastricht, The Netherlands; Department of Experimental Biomedicine, University Hospital and Rudolf Virchow Center, University of Würzburg, Würzburg, Germany; Walter Brendel Centre of Experimental Medicine and German Centre of Cardiovascular Research, Munich Heart Alliance, Ludwig-Maximilians-Universität München, München, Germany; Department of Developmental Biology, Centre for Medical Biotechnology, University of Duisburg-Essen, Duisburg-Essen, Germany; Aragon Institute of Health Sciences I+CS/IIS and ARAID, Zaragoza, Spain; School of Physiology and Pharmacology, University of Bristol, Bristol, United Kingdom; Institute of Physiology, University of Regensburg, Regensburg, Germany; **Arthur Bloom Haemophilia Centre, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - Elisabetta Castoldi
- *Department of Cell Biochemistry of Thrombosis and Haemostasis Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), University of Maastricht, Maastricht, The Netherlands; Department of Experimental Biomedicine, University Hospital and Rudolf Virchow Center, University of Würzburg, Würzburg, Germany; Walter Brendel Centre of Experimental Medicine and German Centre of Cardiovascular Research, Munich Heart Alliance, Ludwig-Maximilians-Universität München, München, Germany; Department of Developmental Biology, Centre for Medical Biotechnology, University of Duisburg-Essen, Duisburg-Essen, Germany; Aragon Institute of Health Sciences I+CS/IIS and ARAID, Zaragoza, Spain; School of Physiology and Pharmacology, University of Bristol, Bristol, United Kingdom; Institute of Physiology, University of Regensburg, Regensburg, Germany; **Arthur Bloom Haemophilia Centre, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - Joachim Pircher
- *Department of Cell Biochemistry of Thrombosis and Haemostasis Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), University of Maastricht, Maastricht, The Netherlands; Department of Experimental Biomedicine, University Hospital and Rudolf Virchow Center, University of Würzburg, Würzburg, Germany; Walter Brendel Centre of Experimental Medicine and German Centre of Cardiovascular Research, Munich Heart Alliance, Ludwig-Maximilians-Universität München, München, Germany; Department of Developmental Biology, Centre for Medical Biotechnology, University of Duisburg-Essen, Duisburg-Essen, Germany; Aragon Institute of Health Sciences I+CS/IIS and ARAID, Zaragoza, Spain; School of Physiology and Pharmacology, University of Bristol, Bristol, United Kingdom; Institute of Physiology, University of Regensburg, Regensburg, Germany; **Arthur Bloom Haemophilia Centre, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - Constance C F M J Baaten
- *Department of Cell Biochemistry of Thrombosis and Haemostasis Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), University of Maastricht, Maastricht, The Netherlands; Department of Experimental Biomedicine, University Hospital and Rudolf Virchow Center, University of Würzburg, Würzburg, Germany; Walter Brendel Centre of Experimental Medicine and German Centre of Cardiovascular Research, Munich Heart Alliance, Ludwig-Maximilians-Universität München, München, Germany; Department of Developmental Biology, Centre for Medical Biotechnology, University of Duisburg-Essen, Duisburg-Essen, Germany; Aragon Institute of Health Sciences I+CS/IIS and ARAID, Zaragoza, Spain; School of Physiology and Pharmacology, University of Bristol, Bristol, United Kingdom; Institute of Physiology, University of Regensburg, Regensburg, Germany; **Arthur Bloom Haemophilia Centre, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - Manuela Wülling
- *Department of Cell Biochemistry of Thrombosis and Haemostasis Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), University of Maastricht, Maastricht, The Netherlands; Department of Experimental Biomedicine, University Hospital and Rudolf Virchow Center, University of Würzburg, Würzburg, Germany; Walter Brendel Centre of Experimental Medicine and German Centre of Cardiovascular Research, Munich Heart Alliance, Ludwig-Maximilians-Universität München, München, Germany; Department of Developmental Biology, Centre for Medical Biotechnology, University of Duisburg-Essen, Duisburg-Essen, Germany; Aragon Institute of Health Sciences I+CS/IIS and ARAID, Zaragoza, Spain; School of Physiology and Pharmacology, University of Bristol, Bristol, United Kingdom; Institute of Physiology, University of Regensburg, Regensburg, Germany; **Arthur Bloom Haemophilia Centre, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - Marijke J E Kuijpers
- *Department of Cell Biochemistry of Thrombosis and Haemostasis Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), University of Maastricht, Maastricht, The Netherlands; Department of Experimental Biomedicine, University Hospital and Rudolf Virchow Center, University of Würzburg, Würzburg, Germany; Walter Brendel Centre of Experimental Medicine and German Centre of Cardiovascular Research, Munich Heart Alliance, Ludwig-Maximilians-Universität München, München, Germany; Department of Developmental Biology, Centre for Medical Biotechnology, University of Duisburg-Essen, Duisburg-Essen, Germany; Aragon Institute of Health Sciences I+CS/IIS and ARAID, Zaragoza, Spain; School of Physiology and Pharmacology, University of Bristol, Bristol, United Kingdom; Institute of Physiology, University of Regensburg, Regensburg, Germany; **Arthur Bloom Haemophilia Centre, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - Ralf Köhler
- *Department of Cell Biochemistry of Thrombosis and Haemostasis Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), University of Maastricht, Maastricht, The Netherlands; Department of Experimental Biomedicine, University Hospital and Rudolf Virchow Center, University of Würzburg, Würzburg, Germany; Walter Brendel Centre of Experimental Medicine and German Centre of Cardiovascular Research, Munich Heart Alliance, Ludwig-Maximilians-Universität München, München, Germany; Department of Developmental Biology, Centre for Medical Biotechnology, University of Duisburg-Essen, Duisburg-Essen, Germany; Aragon Institute of Health Sciences I+CS/IIS and ARAID, Zaragoza, Spain; School of Physiology and Pharmacology, University of Bristol, Bristol, United Kingdom; Institute of Physiology, University of Regensburg, Regensburg, Germany; **Arthur Bloom Haemophilia Centre, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - Alastair W Poole
- *Department of Cell Biochemistry of Thrombosis and Haemostasis Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), University of Maastricht, Maastricht, The Netherlands; Department of Experimental Biomedicine, University Hospital and Rudolf Virchow Center, University of Würzburg, Würzburg, Germany; Walter Brendel Centre of Experimental Medicine and German Centre of Cardiovascular Research, Munich Heart Alliance, Ludwig-Maximilians-Universität München, München, Germany; Department of Developmental Biology, Centre for Medical Biotechnology, University of Duisburg-Essen, Duisburg-Essen, Germany; Aragon Institute of Health Sciences I+CS/IIS and ARAID, Zaragoza, Spain; School of Physiology and Pharmacology, University of Bristol, Bristol, United Kingdom; Institute of Physiology, University of Regensburg, Regensburg, Germany; **Arthur Bloom Haemophilia Centre, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - Rainer Schreiber
- *Department of Cell Biochemistry of Thrombosis and Haemostasis Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), University of Maastricht, Maastricht, The Netherlands; Department of Experimental Biomedicine, University Hospital and Rudolf Virchow Center, University of Würzburg, Würzburg, Germany; Walter Brendel Centre of Experimental Medicine and German Centre of Cardiovascular Research, Munich Heart Alliance, Ludwig-Maximilians-Universität München, München, Germany; Department of Developmental Biology, Centre for Medical Biotechnology, University of Duisburg-Essen, Duisburg-Essen, Germany; Aragon Institute of Health Sciences I+CS/IIS and ARAID, Zaragoza, Spain; School of Physiology and Pharmacology, University of Bristol, Bristol, United Kingdom; Institute of Physiology, University of Regensburg, Regensburg, Germany; **Arthur Bloom Haemophilia Centre, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - Andrea Vortkamp
- *Department of Cell Biochemistry of Thrombosis and Haemostasis Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), University of Maastricht, Maastricht, The Netherlands; Department of Experimental Biomedicine, University Hospital and Rudolf Virchow Center, University of Würzburg, Würzburg, Germany; Walter Brendel Centre of Experimental Medicine and German Centre of Cardiovascular Research, Munich Heart Alliance, Ludwig-Maximilians-Universität München, München, Germany; Department of Developmental Biology, Centre for Medical Biotechnology, University of Duisburg-Essen, Duisburg-Essen, Germany; Aragon Institute of Health Sciences I+CS/IIS and ARAID, Zaragoza, Spain; School of Physiology and Pharmacology, University of Bristol, Bristol, United Kingdom; Institute of Physiology, University of Regensburg, Regensburg, Germany; **Arthur Bloom Haemophilia Centre, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - Peter W Collins
- *Department of Cell Biochemistry of Thrombosis and Haemostasis Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), University of Maastricht, Maastricht, The Netherlands; Department of Experimental Biomedicine, University Hospital and Rudolf Virchow Center, University of Würzburg, Würzburg, Germany; Walter Brendel Centre of Experimental Medicine and German Centre of Cardiovascular Research, Munich Heart Alliance, Ludwig-Maximilians-Universität München, München, Germany; Department of Developmental Biology, Centre for Medical Biotechnology, University of Duisburg-Essen, Duisburg-Essen, Germany; Aragon Institute of Health Sciences I+CS/IIS and ARAID, Zaragoza, Spain; School of Physiology and Pharmacology, University of Bristol, Bristol, United Kingdom; Institute of Physiology, University of Regensburg, Regensburg, Germany; **Arthur Bloom Haemophilia Centre, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - Bernhard Nieswandt
- *Department of Cell Biochemistry of Thrombosis and Haemostasis Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), University of Maastricht, Maastricht, The Netherlands; Department of Experimental Biomedicine, University Hospital and Rudolf Virchow Center, University of Würzburg, Würzburg, Germany; Walter Brendel Centre of Experimental Medicine and German Centre of Cardiovascular Research, Munich Heart Alliance, Ludwig-Maximilians-Universität München, München, Germany; Department of Developmental Biology, Centre for Medical Biotechnology, University of Duisburg-Essen, Duisburg-Essen, Germany; Aragon Institute of Health Sciences I+CS/IIS and ARAID, Zaragoza, Spain; School of Physiology and Pharmacology, University of Bristol, Bristol, United Kingdom; Institute of Physiology, University of Regensburg, Regensburg, Germany; **Arthur Bloom Haemophilia Centre, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - Karl Kunzelmann
- *Department of Cell Biochemistry of Thrombosis and Haemostasis Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), University of Maastricht, Maastricht, The Netherlands; Department of Experimental Biomedicine, University Hospital and Rudolf Virchow Center, University of Würzburg, Würzburg, Germany; Walter Brendel Centre of Experimental Medicine and German Centre of Cardiovascular Research, Munich Heart Alliance, Ludwig-Maximilians-Universität München, München, Germany; Department of Developmental Biology, Centre for Medical Biotechnology, University of Duisburg-Essen, Duisburg-Essen, Germany; Aragon Institute of Health Sciences I+CS/IIS and ARAID, Zaragoza, Spain; School of Physiology and Pharmacology, University of Bristol, Bristol, United Kingdom; Institute of Physiology, University of Regensburg, Regensburg, Germany; **Arthur Bloom Haemophilia Centre, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - Judith M E M Cosemans
- *Department of Cell Biochemistry of Thrombosis and Haemostasis Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), University of Maastricht, Maastricht, The Netherlands; Department of Experimental Biomedicine, University Hospital and Rudolf Virchow Center, University of Würzburg, Würzburg, Germany; Walter Brendel Centre of Experimental Medicine and German Centre of Cardiovascular Research, Munich Heart Alliance, Ludwig-Maximilians-Universität München, München, Germany; Department of Developmental Biology, Centre for Medical Biotechnology, University of Duisburg-Essen, Duisburg-Essen, Germany; Aragon Institute of Health Sciences I+CS/IIS and ARAID, Zaragoza, Spain; School of Physiology and Pharmacology, University of Bristol, Bristol, United Kingdom; Institute of Physiology, University of Regensburg, Regensburg, Germany; **Arthur Bloom Haemophilia Centre, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - Johan W M Heemskerk
- *Department of Cell Biochemistry of Thrombosis and Haemostasis Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), University of Maastricht, Maastricht, The Netherlands; Department of Experimental Biomedicine, University Hospital and Rudolf Virchow Center, University of Würzburg, Würzburg, Germany; Walter Brendel Centre of Experimental Medicine and German Centre of Cardiovascular Research, Munich Heart Alliance, Ludwig-Maximilians-Universität München, München, Germany; Department of Developmental Biology, Centre for Medical Biotechnology, University of Duisburg-Essen, Duisburg-Essen, Germany; Aragon Institute of Health Sciences I+CS/IIS and ARAID, Zaragoza, Spain; School of Physiology and Pharmacology, University of Bristol, Bristol, United Kingdom; Institute of Physiology, University of Regensburg, Regensburg, Germany; **Arthur Bloom Haemophilia Centre, School of Medicine, Cardiff University, Cardiff, United Kingdom
| |
Collapse
|
31
|
Brooks MB, Catalfamo JL, MacNguyen R, Tim D, Fancher S, McCardle JA. A TMEM16F point mutation causes an absence of canine platelet TMEM16F and ineffective activation and death-induced phospholipid scrambling. J Thromb Haemost 2015; 13:2240-52. [PMID: 26414452 DOI: 10.1111/jth.13157] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2015] [Accepted: 09/12/2015] [Indexed: 12/24/2022]
Abstract
BACKGROUND TMEM16F is an ion channel and calcium-dependent lipid scramblase that mediates phosphatidylserine (PS) exposure on the plasma membrane. Two disparate disease phenotypes are associated with TMEM16F loss-of-function mutations: a rare bleeding disorder (Scott syndrome) and skeletal malformations due to aberrant bone mineralization in a TMEM16F knockout mouse. We therefore undertook comparative studies of TMEM16F expression in canine Scott syndrome (CSS), an autosomal recessive platelet defect. OBJECTIVES To define anoctamin proteins and scramblase response of CSS platelets and to determine whether TMEM16F is the CSS disease gene. METHODS CSS TMEM16F cDNA and gene were sequenced and mutation detection was performed in CSS pedigrees. Platelet fractions from CSS dogs were isolated for proteomic and immunologic characterization of TMEM16F. Annexin V was used as a flow cytometric marker of induced platelet PS externalization. RESULTS A TMEM16F splice site mutation segregated with the CSS trait and TMEM16F protein was undetectable in CSS platelet membranes; however, a second anoctamin, TMEM16K, was found. Proteomic analyses revealed a network of 32 proteins that differentially cosegregated with platelet plasma membrane TMEM16F. CSS platelets had profoundly impaired scramblase response to pharmacologic and physiologic agents that increase intraplatelet calcium and conditions that induce apoptotic and necrotic cell death. CONCLUSIONS CSS platelets represent a TMEM16F-null mutant model that demonstrates a central role for TMEM16F in mediating platelet PS externalization in response to activating and death signals. Platelet TMEM16F may prove a novel drug target for modulating platelet procoagulant activity and extending platelet life span.
Collapse
Affiliation(s)
- M B Brooks
- Department of Population Medicine and Diagnostic Sciences, College of Veterinary Medicine, Cornell University, Ithaca, NY, USA
| | - J L Catalfamo
- Department of Population Medicine and Diagnostic Sciences, College of Veterinary Medicine, Cornell University, Ithaca, NY, USA
| | - R MacNguyen
- Department of Population Medicine and Diagnostic Sciences, College of Veterinary Medicine, Cornell University, Ithaca, NY, USA
| | - D Tim
- Department of Population Medicine and Diagnostic Sciences, College of Veterinary Medicine, Cornell University, Ithaca, NY, USA
| | - S Fancher
- Department of Population Medicine and Diagnostic Sciences, College of Veterinary Medicine, Cornell University, Ithaca, NY, USA
| | - J A McCardle
- Institute of Biotechnology, Cornell University, Ithaca, NY, USA
| |
Collapse
|
32
|
Hoffmann EK, Sørensen BH, Sauter DPR, Lambert IH. Role of volume-regulated and calcium-activated anion channels in cell volume homeostasis, cancer and drug resistance. Channels (Austin) 2015; 9:380-96. [PMID: 26569161 DOI: 10.1080/19336950.2015.1089007] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022] Open
Abstract
Volume-regulated channels for anions (VRAC) / organic osmolytes (VSOAC) play essential roles in cell volume regulation and other cellular functions, e.g. proliferation, cell migration and apoptosis. LRRC8A, which belongs to the leucine rich-repeat containing protein family, was recently shown to be an essential component of both VRAC and VSOAC. Reduced VRAC and VSOAC activities are seen in drug resistant cancer cells. ANO1 is a calcium-activated chloride channel expressed on the plasma membrane of e.g., secretory epithelia. ANO1 is amplified and highly expressed in a large number of carcinomas. The gene, encoding for ANO1, maps to a region on chromosome 11 (11q13) that is frequently amplified in cancer cells. Knockdown of ANO1 impairs cell proliferation and cell migration in several cancer cells. Below we summarize the basic biophysical properties of VRAC, VSOAC and ANO1 and their most important cellular functions as well as their role in cancer and drug resistance.
Collapse
Affiliation(s)
- Else K Hoffmann
- a Department of Biology ; Section for Cell Biology and Physiology; University of Copenhagen ; Copenhagen , Denmark
| | - Belinda H Sørensen
- a Department of Biology ; Section for Cell Biology and Physiology; University of Copenhagen ; Copenhagen , Denmark
| | - Daniel P R Sauter
- a Department of Biology ; Section for Cell Biology and Physiology; University of Copenhagen ; Copenhagen , Denmark
| | - Ian H Lambert
- a Department of Biology ; Section for Cell Biology and Physiology; University of Copenhagen ; Copenhagen , Denmark
| |
Collapse
|
33
|
Rysavy NM, Shimoda LMN, Dixon AM, Speck M, Stokes AJ, Turner H, Umemoto EY. Beyond apoptosis: the mechanism and function of phosphatidylserine asymmetry in the membrane of activating mast cells. BIOARCHITECTURE 2015; 4:127-37. [PMID: 25759911 DOI: 10.1080/19490992.2014.995516] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Loss of plasma membrane asymmetry is a hallmark of apoptosis, but lipid bilayer asymmetry and loss of asymmetry can contribute to numerous cellular functions and responses that are independent of programmed cell death. Exofacial exposure of phosphatidylserine occurs in lymphocytes and mast cells after antigenic stimulation and in the absence of apoptosis, suggesting that there is a functional requirement for phosphatidylserine exposure in immunocytes. In this review we examine current ideas as to the nature of this functional role in mast cell activation. Mechanistically, there is controversy as to the candidate proteins responsible for phosphatidylserine translocation from the internal to external leaflet, and here we review the candidacies of mast cell PLSCR1 and TMEM16F. Finally we examine the potential relationship between functionally important mast cell membrane perturbations and phosphatidylserine exposure during activation.
Collapse
Key Words
- ABCA, ABC binding cassette family A
- CRAC, calcium release activated channel
- GPMV, giant plasma membrane vesicle
- ITIM, immunoreceptor tyrosine based inhibitory motif
- PLA2, phospholipase A2
- PLSCR, phospholipid scramblase
- PMA, phorbol 12,13-myristate acetate
- RBL, rat basophilic leukemia
- RFU, relative fluorescence units
- ROI, region of interest
- TMEM, transmembrane protein
- TMEM16F
- WGA, wheat germ agglutinin
- mast cells
- membrane lipids
- phosphatidylserine
Collapse
Affiliation(s)
- Noel M Rysavy
- a Laboratory of Immunology and Signal Transduction ; Department of Biology; Chaminade University ; Honolulu , Hawai'i USA
| | | | | | | | | | | | | |
Collapse
|
34
|
Forschbach V, Goppelt-Struebe M, Kunzelmann K, Schreiber R, Piedagnel R, Kraus A, Eckardt KU, Buchholz B. Anoctamin 6 is localized in the primary cilium of renal tubular cells and is involved in apoptosis-dependent cyst lumen formation. Cell Death Dis 2015; 6:e1899. [PMID: 26448322 PMCID: PMC4632301 DOI: 10.1038/cddis.2015.273] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2015] [Revised: 08/11/2015] [Accepted: 08/27/2015] [Indexed: 02/07/2023]
Abstract
Primary cilia are antenna-like structures projected from the apical surface of various mammalian cells including renal tubular cells. Functional or structural defects of the cilium lead to systemic disorders comprising polycystic kidneys as a key feature. Here we show that anoctamin 6 (ANO6), a member of the anoctamin chloride channel family, is localized in the primary cilium of renal epithelial cells in vitro and in vivo. ANO6 was not essential for cilia formation and had no effect on in vitro cyst expansion. However, knockdown of ANO6 impaired cyst lumen formation of MDCK cells in three-dimensional culture. In the absence of ANO6, apoptosis was reduced and epithelial cells were incompletely removed from the center of cell aggregates, which form in the early phase of cystogenesis. In line with these data, we show that ANO6 is highly expressed in apoptotic cyst epithelial cells of human polycystic kidneys. These data identify ANO6 as a cilium-associated protein and suggest its functional relevance in cyst formation.
Collapse
Affiliation(s)
- V Forschbach
- Department of Nephrology and Hypertension, Friedrich-Alexander-University Erlangen-Nuernberg, 91054 Erlangen, Germany
| | - M Goppelt-Struebe
- Department of Nephrology and Hypertension, Friedrich-Alexander-University Erlangen-Nuernberg, 91054 Erlangen, Germany
| | - K Kunzelmann
- Department of Physiology, University of Regensburg, 93053 Regensburg, Germany
| | - R Schreiber
- Department of Physiology, University of Regensburg, 93053 Regensburg, Germany
| | - R Piedagnel
- Sorbonne Universités, UPMC Univ Paris 06, UMR_S 1155, F-75005 Paris, France
- INSERM, UMR_S 1155, F-75005 Paris, France
| | - A Kraus
- Department of Nephrology and Hypertension, Friedrich-Alexander-University Erlangen-Nuernberg, 91054 Erlangen, Germany
| | - K-U Eckardt
- Department of Nephrology and Hypertension, Friedrich-Alexander-University Erlangen-Nuernberg, 91054 Erlangen, Germany
| | - B Buchholz
- Department of Nephrology and Hypertension, Friedrich-Alexander-University Erlangen-Nuernberg, 91054 Erlangen, Germany
| |
Collapse
|
35
|
Cellular volume regulation by anoctamin 6: Ca²⁺, phospholipase A2 and osmosensing. Pflugers Arch 2015; 468:335-49. [PMID: 26438191 DOI: 10.1007/s00424-015-1739-8] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2015] [Revised: 08/30/2015] [Accepted: 09/29/2015] [Indexed: 02/08/2023]
Abstract
During cell swelling, Cl(-) channels are activated to lower intracellular Cl(-) concentrations and to reduce cell volume, a process termed regulatory volume decrease (RVD). We show that anoctamin 6 (ANO6; TMEM16F) produces volume-regulated anion currents and controls cell volume in four unrelated cell types. Volume regulation is compromised in freshly isolated intestinal epithelial cells from Ano6-/- mice and also in lymphocytes from a patient lacking expression of ANO6. Ca(2+) influx is activated and thus ANO6 is stimulated during cell swelling by local Ca(2+) increase probably in functional nanodomains near the plasma membrane. This leads to stimulation of phospholipase A2 (PLA2) and generation of plasma membrane lysophospholipids, which activates ANO6. Direct application of lysophospholipids also activates an anion current that is inhibited by typical ANO6 blocker. An increase in intracellular Ca(2+) supports activation of ANO6, but is not required when PLA2 is fully activated, while re-addition of arachidonic acid completely blocked ANO6. Moreover, ANO6 is activated by low intracellular Cl(-) concentrations and may therefore operate as a cellular osmosensor. High intracellular Cl(-) concentration inhibits ANO6 and activation by PLA2. Taken together, ANO6 supports volume regulation and volume activation of anion currents by action as a Cl(-) channel or by scrambling membrane phospholipids. Thereby, it may support the function of LRRC8 proteins.
Collapse
|
36
|
Gasperi V, Avigliano L, Evangelista D, Oddi S, Chiurchiù V, Lanuti M, Maccarrone M, Valeria Catani M. 2-Arachidonoylglycerol enhances platelet formation from human megakaryoblasts. Cell Cycle 2015; 13:3938-47. [PMID: 25427281 DOI: 10.4161/15384101.2014.982941] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Platelets modulate vascular system integrity, and their loss is critical in haematological pathologies and after chemotherapy. Therefore, identification of molecules enhancing platelet production would be useful to counteract thrombocytopenia. We have previously shown that 2-arachidonoylglycerol (2-AG) acts as a true agonist of platelets, as well as it commits erythroid precursors toward the megakaryocytic lineage. Against this background, we sought to further interrogate the role of 2-AG in megakaryocyte/platelet physiology by investigating terminal differentiation, and subsequent thrombopoiesis. To this end, we used MEG-01 cells, a human megakaryoblastic cell line able to produce in vitro platelet-like particles. 2-AG increased the number of cells showing ruffled surface and enhanced surface expression of specific megakaryocyte/platelet surface antigens, typical hallmarks of terminal megakaryocytic differentiation and platelet production. Changes in cytoskeleton modeling also occurred in differentiated megakaryocytes and blebbing platelets. 2-AG acted by binding to CB1 and CB2 receptors, because specific antagonists reverted its effect. Platelets were split off from megakaryocytes and were functional: they contained the platelet-specific surface markers CD61 and CD49, whose levels increased following stimulation with a natural agonist like collagen. Given the importance of 2-AG for driving megakaryopoiesis and thrombopoiesis, not surprisingly we found that its hydrolytic enzymes were tightly controlled by classical inducers of megakaryocyte differentiation. In conclusion 2-AG, by triggering megakaryocyte maturation and platelet release, may have clinical efficacy to counteract thrombocytopenia-related diseases.
Collapse
Key Words
- 2-AG, 2-arachidonoylglycerol
- AEA, anandamide
- APC, allophycocyanin
- CB1, type-1 cannabinoid receptor
- CB2, type-2 cannabinoid receptor
- CD, cluster of differentiation
- DAGL, diacylglycerol lipase
- Differentiation
- FAAH, fatty acid amide hydrolase
- FITC, fluorescein isothiocyanate
- HEL, human erythroleukemia
- MAGL, monoacylglycerol lipase
- PE, phycoerythrin
- TPA, 12-O-tetradecanoylphorbol-13-acetate
- cluster of differentiation
- cytoskeleton
- eCB, endocannabinoid
- endocannabinoid system
- haematopoietic cells
- megakaryocytes
- platelets
Collapse
Affiliation(s)
- Valeria Gasperi
- a Department of Experimental Medicine & Surgery ; University of Rome Tor Vergata ; Rome , Italy
| | | | | | | | | | | | | | | |
Collapse
|
37
|
TMEM16, LRRC8A, bestrophin: chloride channels controlled by Ca2+ and cell volume. Trends Biochem Sci 2015; 40:535-43. [DOI: 10.1016/j.tibs.2015.07.005] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2015] [Revised: 07/10/2015] [Accepted: 07/10/2015] [Indexed: 01/13/2023]
|
38
|
Henne WM, Liou J, Emr SD. Molecular mechanisms of inter-organelle ER–PM contact sites. Curr Opin Cell Biol 2015; 35:123-30. [DOI: 10.1016/j.ceb.2015.05.001] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2015] [Revised: 04/23/2015] [Accepted: 05/01/2015] [Indexed: 10/23/2022]
|
39
|
Yu K, Whitlock JM, Lee K, Ortlund EA, Yuan Cui Y, Hartzell HC. Identification of a lipid scrambling domain in ANO6/TMEM16F. eLife 2015; 4:e06901. [PMID: 26057829 PMCID: PMC4477620 DOI: 10.7554/elife.06901] [Citation(s) in RCA: 138] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2015] [Accepted: 06/08/2015] [Indexed: 12/22/2022] Open
Abstract
Phospholipid scrambling (PLS) is a ubiquitous cellular mechanism involving the regulated bidirectional transport of phospholipids down their concentration gradient between membrane leaflets. ANO6/TMEM16F has been shown to be essential for Ca(2+)-dependent PLS, but controversy surrounds whether ANO6 is a phospholipid scramblase or an ion channel like other ANO/TMEM16 family members. Combining patch clamp recording with measurement of PLS, we show that ANO6 elicits robust Ca(2+)-dependent PLS coinciding with ionic currents that are explained by ionic leak during phospholipid translocation. By analyzing ANO1-ANO6 chimeric proteins, we identify a domain in ANO6 necessary for PLS and sufficient to confer this function on ANO1, which normally does not scramble. Homology modeling shows that the scramblase domain forms an unusual hydrophilic cleft that faces the lipid bilayer and may function to facilitate translocation of phospholipid between membrane leaflets. These findings provide a mechanistic framework for understanding PLS and how ANO6 functions in this process.
Collapse
Affiliation(s)
- Kuai Yu
- Department of Cell Biology, Emory University School of Medicine, Atlanta, United States
| | - Jarred M Whitlock
- Department of Cell Biology, Emory University School of Medicine, Atlanta, United States
| | - Kyleen Lee
- Department of Cell Biology, Emory University School of Medicine, Atlanta, United States
| | - Eric A Ortlund
- Department of Cell Biology, Emory University School of Medicine, Atlanta, United States
- Department of Biochemistry, Emory University School of Medicine, Atlanta, United States
| | - Yuan Yuan Cui
- Department of Cell Biology, Emory University School of Medicine, Atlanta, United States
| | - H Criss Hartzell
- Department of Cell Biology, Emory University School of Medicine, Atlanta, United States
| |
Collapse
|
40
|
Ousingsawat J, Wanitchakool P, Kmit A, Romao AM, Jantarajit W, Schreiber R, Kunzelmann K. Anoctamin 6 mediates effects essential for innate immunity downstream of P2X7 receptors in macrophages. Nat Commun 2015; 6:6245. [PMID: 25651887 DOI: 10.1038/ncomms7245] [Citation(s) in RCA: 121] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2014] [Accepted: 01/08/2015] [Indexed: 12/18/2022] Open
Abstract
Purinergic P2X7 receptors (P2X7R) are fundamental to innate immune response. In macrophages, transient stimulation of P2X7R activates several transport mechanisms and induces the scrambling of phospholipids with subsequent membrane blebbing and apoptosis. These processes support phagocytosis and subsequent killing of phagocytosed bacteria. Here we demonstrate that the stimulation of P2X7 receptors activates anoctamin 6 (ANO6, TMEM16F), a protein that functions as Ca(2+) dependent phospholipid scramblase and Ca(2+)-activated Cl(-) channel. Inhibition or knockdown of ANO6 attenuates ATP-induced cell shrinkage, cell migration and phospholipid scrambling. In mouse macrophages, Ano6 produces large ion currents by stimulation of P2X7 receptors and contributes to ATP-induced membrane blebbing and apoptosis, which is largely reduced in macrophages from Ano6-/- mice. ANO6 supports bacterial phagocytosis and killing by mouse and human THP-1 macrophages. Our data demonstrate that anoctamin 6 is an essential component of the immune defense by macrophages.
Collapse
Affiliation(s)
- Jiraporn Ousingsawat
- Physiological Institute, University of Regensburg, Universitätsstraße 31, D-93053 Regensburg, Germany
| | - Podchanart Wanitchakool
- Physiological Institute, University of Regensburg, Universitätsstraße 31, D-93053 Regensburg, Germany
| | - Arthur Kmit
- Physiological Institute, University of Regensburg, Universitätsstraße 31, D-93053 Regensburg, Germany
| | - Ana M Romao
- Physiological Institute, University of Regensburg, Universitätsstraße 31, D-93053 Regensburg, Germany
| | - Walailak Jantarajit
- Physiological Institute, University of Regensburg, Universitätsstraße 31, D-93053 Regensburg, Germany
| | - Rainer Schreiber
- Physiological Institute, University of Regensburg, Universitätsstraße 31, D-93053 Regensburg, Germany
| | - Karl Kunzelmann
- Physiological Institute, University of Regensburg, Universitätsstraße 31, D-93053 Regensburg, Germany
| |
Collapse
|
41
|
Stolz M, Klapperstück M, Kendzierski T, Detro-Dassen S, Panning A, Schmalzing G, Markwardt F. Homodimeric anoctamin-1, but not homodimeric anoctamin-6, is activated by calcium increases mediated by the P2Y1 and P2X7 receptors. Pflugers Arch 2015; 467:2121-40. [PMID: 25592660 DOI: 10.1007/s00424-015-1687-3] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2014] [Revised: 12/23/2014] [Accepted: 01/06/2015] [Indexed: 01/14/2023]
Abstract
The P2X7 receptor (P2X7R) is a ligand-gated ion channel that conducts Na(+), K(+), and Ca(2+) when activated by extracellular ATP. In various cell types, such as secretory epithelia, the P2X7R is co-expressed with Ca(2+)-dependent Cl(-) channels of the TMEM16/anoctamin family. Here, we studied whether the P2X7R and TMEM16A/anoctamin-1 (Ano1) or TMEM16F/anoctamin-6 (Ano6) interact functionally and physically, using oocytes of Xenopus laevis and Ambystoma mexicanum (Axolotl) for heterologous expression. As a control, we co-expressed anoctamin-1 with the P2Y1 receptor (P2Y1R), which induces the release of Ca(2+) from intracellular stores via activating phospholipase C through coupling to Gαq. We found that co-expression of anoctamin-1 with the P2Y1R resulted in a small transient increase in Cl(-) conductance in response to ATP. Co-expression of anoctamin-1 with the P2X7R resulted in a large sustained increase in Cl(-) conductance via Ca(2+) influx through the ATP-opened P2X7R in Xenopus and in Axolotl oocytes, which lack endogenous Ca(2+)-dependent Cl(-) channels. P2Y1R- or P2X7R-mediated stimulation of Ano1 was primarily functional, as demonstrated by the absence of a physically stable interaction between Ano1 and the P2X7R. In the pancreatic cell line AsPC-1, we found the same functional Ca(2+)-dependent interaction of P2X7R and Ano1. The P2X7R-mediated sustained activation of Ano1 may be physiologically relevant to the time course of stimulus-secretion coupling in secretory epithelia. No such increase in Cl(-) conductance could be elicited by activating the P2X7 receptor in either Xenopus oocytes or Axolotl oocytes co-expressing Ano6. The lack of function of Ano6 can, at least in part, be explained by its poor cell-surface expression, resulting from a relatively inefficient exit of the homodimeric Ano6 from the endoplasmic reticulum.
Collapse
Affiliation(s)
- Michaela Stolz
- Molecular Pharmacology, RWTH Aachen University, Wendlingweg 2, D-52074, Aachen, Germany
| | - Manuela Klapperstück
- Julius-Bernstein-Institute for Physiology, Martin-Luther-University Halle-Wittenberg, Magdeburger Str. 6, D-06097, Halle/Saale, Germany
| | - Thomas Kendzierski
- Julius-Bernstein-Institute for Physiology, Martin-Luther-University Halle-Wittenberg, Magdeburger Str. 6, D-06097, Halle/Saale, Germany
| | - Silvia Detro-Dassen
- Molecular Pharmacology, RWTH Aachen University, Wendlingweg 2, D-52074, Aachen, Germany
| | - Anna Panning
- Molecular Pharmacology, RWTH Aachen University, Wendlingweg 2, D-52074, Aachen, Germany
| | - Günther Schmalzing
- Molecular Pharmacology, RWTH Aachen University, Wendlingweg 2, D-52074, Aachen, Germany
| | - Fritz Markwardt
- Julius-Bernstein-Institute for Physiology, Martin-Luther-University Halle-Wittenberg, Magdeburger Str. 6, D-06097, Halle/Saale, Germany.
| |
Collapse
|
42
|
Ousingsawat J, Wanitchakool P, Schreiber R, Wuelling M, Vortkamp A, Kunzelmann K. Anoctamin-6 controls bone mineralization by activating the calcium transporter NCX1. J Biol Chem 2015; 290:6270-80. [PMID: 25589784 DOI: 10.1074/jbc.m114.602979] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Anoctamin-6 (Ano6, TMEM16F) belongs to a family of putative Ca(2+)-activated Cl(-) channels and operates as membrane phospholipid scramblase. Deletion of Ano6 leads to reduced skeleton size, skeletal deformities, and mineralization defects in mice. However, it remains entirely unclear how a lack of Ano6 leads to a delay in bone mineralization by osteoblasts. The Na(+)/Ca(2+) exchanger NCX1 was found to interact with Ano6 in a two-hybrid split-ubiquitin screen. Using human osteoblasts and osteoblasts from Ano6(-/-) and WT mice, we demonstrate that NCX1 requires Ano6 to efficiently translocate Ca(2+) out of osteoblasts into the calcifying bone matrix. Ca(2+)-activated anion currents are missing in primary osteoblasts isolated from Ano6 null mice. Our findings demonstrate the importance of NCX1 for bone mineralization and explain why deletion of an ion channel leads to the observed mineralization defect: Ano6 Cl(-) currents are probably required to operate as a Cl(-) bypass channel, thereby compensating net Na(+) charge movement by NCX1.
Collapse
Affiliation(s)
- Jiraporn Ousingsawat
- From the Institut für Physiologie, Universität Regensburg, D-93053 Regensburg and
| | | | - Rainer Schreiber
- From the Institut für Physiologie, Universität Regensburg, D-93053 Regensburg and
| | - Manuela Wuelling
- the Department Entwicklungsbiologie, Fakultät für Biologie, Universität Duisburg-Essen, 45141 Essen, Germany
| | - Andrea Vortkamp
- the Department Entwicklungsbiologie, Fakultät für Biologie, Universität Duisburg-Essen, 45141 Essen, Germany
| | - Karl Kunzelmann
- From the Institut für Physiologie, Universität Regensburg, D-93053 Regensburg and
| |
Collapse
|
43
|
Henkel B, Drose DR, Ackels T, Oberland S, Spehr M, Neuhaus EM. Co-expression of anoctamins in cilia of olfactory sensory neurons. Chem Senses 2014; 40:73-87. [PMID: 25500808 DOI: 10.1093/chemse/bju061] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Vertebrates can sense and identify a vast array of chemical cues. The molecular machinery involved in chemodetection and transduction is expressed within the cilia of olfactory sensory neurons. Currently, there is only limited information available on the distribution and density of individual signaling components within the ciliary compartment. Using super-resolution microscopy, we show here that cyclic-nucleotide-gated channels and calcium-activated chloride channels of the anoctamin family are localized to discrete microdomains in the ciliary membrane. In addition to ANO2, a second anoctamin, ANO6, also localizes to ciliary microdomains. This observation, together with the fact that ANO6 and ANO2 co-localize, indicates a role for ANO6 in olfactory signaling. We show that both ANO2 and ANO6 can form heteromultimers and that this heteromerization alters the recombinant channels' physiological properties. Thus, we provide evidence for interaction of ANO2 and ANO6 in olfactory cilia, with possible physiological relevance for olfactory signaling.
Collapse
Affiliation(s)
- Bastian Henkel
- Department of Pharmacology and Toxicology, University Hospital Jena, Drackendorfer Strasse 1, 07747 Jena, Germany, Cluster of Excellence NeuroCure, Charité-Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany, FU Berlin, Fachbereich Biologie, Chemie und Pharmazie , Takustr. 3, 14195 Berlin, Germany and
| | - Daniela R Drose
- Department of Chemosensation, Institute for Biology II, RWTH Aachen University, Worringerweg 3, 52074 Aachen, Germany
| | - Tobias Ackels
- Department of Chemosensation, Institute for Biology II, RWTH Aachen University, Worringerweg 3, 52074 Aachen, Germany
| | - Sonja Oberland
- Department of Pharmacology and Toxicology, University Hospital Jena, Drackendorfer Strasse 1, 07747 Jena, Germany, Cluster of Excellence NeuroCure, Charité-Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany, FU Berlin, Fachbereich Biologie, Chemie und Pharmazie , Takustr. 3, 14195 Berlin, Germany and
| | - Marc Spehr
- Department of Chemosensation, Institute for Biology II, RWTH Aachen University, Worringerweg 3, 52074 Aachen, Germany
| | - Eva M Neuhaus
- Department of Pharmacology and Toxicology, University Hospital Jena, Drackendorfer Strasse 1, 07747 Jena, Germany, Cluster of Excellence NeuroCure, Charité-Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany,
| |
Collapse
|
44
|
Kodigepalli KM, Bowers K, Sharp A, Nanjundan M. Roles and regulation of phospholipid scramblases. FEBS Lett 2014; 589:3-14. [PMID: 25479087 DOI: 10.1016/j.febslet.2014.11.036] [Citation(s) in RCA: 85] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2014] [Revised: 11/22/2014] [Accepted: 11/24/2014] [Indexed: 02/06/2023]
Abstract
Phospholipid scramblase activity is involved in the collapse of phospholipid (PL) asymmetry at the plasma membrane leading to externalization of phosphatidylserine. This activity is crucial for initiation of the blood coagulation cascade and for recognition/elimination of apoptotic cells by macrophages. Efforts to identify gene products associated with this activity led to the characterization of PL scramblase (PLSCR) and XKR family members which contribute to phosphatidylserine exposure in response to apoptotic stimuli. Meanwhile, TMEM16 family members were identified to externalize phosphatidylserine in response to elevated calcium in Scott syndrome platelets, which is critical for activation of the coagulation cascade. Herein, we report their mechanisms of gene regulation, molecular functions independent of their scrambling activity, and their potential roles in pathogenic conditions.
Collapse
Affiliation(s)
- Karthik M Kodigepalli
- Department of Cell Biology, Microbiology, and Molecular Biology, University of South Florida, Tampa, FL 33620, United States
| | - Kiah Bowers
- Department of Cell Biology, Microbiology, and Molecular Biology, University of South Florida, Tampa, FL 33620, United States
| | - Arielle Sharp
- Department of Cell Biology, Microbiology, and Molecular Biology, University of South Florida, Tampa, FL 33620, United States
| | - Meera Nanjundan
- Department of Cell Biology, Microbiology, and Molecular Biology, University of South Florida, Tampa, FL 33620, United States.
| |
Collapse
|
45
|
Brunner JD, Lim NK, Schenck S, Duerst A, Dutzler R. X-ray structure of a calcium-activated TMEM16 lipid scramblase. Nature 2014; 516:207-12. [DOI: 10.1038/nature13984] [Citation(s) in RCA: 339] [Impact Index Per Article: 30.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2014] [Accepted: 10/20/2014] [Indexed: 12/11/2022]
|
46
|
Hankins HM, Baldridge RD, Xu P, Graham TR. Role of flippases, scramblases and transfer proteins in phosphatidylserine subcellular distribution. Traffic 2014; 16:35-47. [PMID: 25284293 DOI: 10.1111/tra.12233] [Citation(s) in RCA: 212] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2014] [Revised: 09/29/2014] [Accepted: 09/30/2014] [Indexed: 12/11/2022]
Abstract
It is well known that lipids are heterogeneously distributed throughout the cell. Most lipid species are synthesized in the endoplasmic reticulum (ER) and then distributed to different cellular locations in order to create the distinct membrane compositions observed in eukaryotes. However, the mechanisms by which specific lipid species are trafficked to and maintained in specific areas of the cell are poorly understood and constitute an active area of research. Of particular interest is the distribution of phosphatidylserine (PS), an anionic lipid that is enriched in the cytosolic leaflet of the plasma membrane. PS transport occurs by both vesicular and non-vesicular routes, with members of the oxysterol-binding protein family (Osh6 and Osh7) recently implicated in the latter route. In addition, the flippase activity of P4-ATPases helps build PS membrane asymmetry by preferentially translocating PS to the cytosolic leaflet. This asymmetric PS distribution can be used as a signaling device by the regulated activation of scramblases, which rapidly expose PS on the extracellular leaflet and play important roles in blood clotting and apoptosis. This review will discuss recent advances made in the study of phospholipid flippases, scramblases and PS-specific lipid transfer proteins, as well as how these proteins contribute to subcellular PS distribution.
Collapse
Affiliation(s)
- Hannah M Hankins
- Department of Biological Sciences, Vanderbilt University Medical Center, Nashville, TN, 37235, USA
| | | | | | | |
Collapse
|
47
|
TMEM16 proteins: unknown structure and confusing functions. J Mol Biol 2014; 427:94-105. [PMID: 25451786 DOI: 10.1016/j.jmb.2014.09.028] [Citation(s) in RCA: 112] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2014] [Revised: 09/24/2014] [Accepted: 09/29/2014] [Indexed: 11/23/2022]
Abstract
The TMEM16 family of membrane proteins, also known as anoctamins, plays key roles in a variety of physiological functions that range from ion transport to phospholipid scrambling and to regulating other ion channels. The first two family members to be functionally characterized, TMEM16A (ANO1) and TMEM16B (ANO2), form Ca(2+)-activated Cl(-) channels and are important for transepithelial ion transport, olfaction, phototransduction, smooth muscle contraction, nociception, cell proliferation and control of neuronal excitability. The roles of other family members, such as TMEM16C (ANO3), TMEM16D (ANO4), TMEM16F (ANO6), TMEM16G (ANO7) and TMEM16J (ANO9), remain poorly understood and controversial. These homologues were reported to be phospholipid scramblases, ion channels, to have both functions or to be regulatory subunits of other channels. Mutations in TMEM16F cause Scott syndrome, a bleeding disorder caused by impaired Ca(2+)-dependent externalization of phosphatidylserine in activated platelets, suggesting that this homologue might be a scramblase. However, overexpression of TMEM16F has also been associated with a remarkable number of different ion channel types, raising the possibility that this protein might be involved in both ion and lipid transports. The recent identification of an ancestral TMEM16 homologue with intrinsic channel and scramblase activities supports this hypothesis. Thus, the TMEM16 family might have diverged in two or three different subclasses, channels, scramblases and dual-function channel/scramblases. The structural bases and functional implication of such a functional diversity within a single protein family remain to be elucidated and the links between TMEM16 functions and human physiology and pathologies need to be investigated.
Collapse
|
48
|
Anoctamins support calcium-dependent chloride secretion by facilitating calcium signaling in adult mouse intestine. Pflugers Arch 2014; 467:1203-13. [PMID: 24974903 DOI: 10.1007/s00424-014-1559-2] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2014] [Revised: 06/12/2014] [Accepted: 06/17/2014] [Indexed: 12/13/2022]
Abstract
Intestinal epithelial electrolyte secretion is activated by increase in intracellular cAMP or Ca(2+) and opening of apical Cl(-) channels. In infants and young animals, but not in adults, Ca(2+)-activated chloride channels may cause secretory diarrhea during rotavirus infection. While detailed knowledge exists concerning the contribution of cAMP-activated cystic fibrosis transmembrane conductance regulator (CFTR) channels, analysis of the role of Ca(2+)-dependent Cl(-) channels became possible through identification of the anoctamin (TMEM16) family of proteins. We demonstrate expression of several anoctamin paralogues in mouse small and large intestines. Using intestinal-specific mouse knockout models for anoctamin 1 (Ano1) and anoctamin 10 (Ano10) and a conventional knockout model for anoctamin 6 (Ano6), we demonstrate the role of anoctamins for Ca(2+)-dependent Cl(-) secretion induced by the muscarinic agonist carbachol (CCH). Ano1 is preferentially expressed in the ileum and large intestine, where it supports Ca(2+)-activated Cl(-) secretion. In contrast, Ano10 is essential for Ca(2+)-dependent Cl(-) secretion in jejunum, where expression of Ano1 was not detected. Although broadly expressed, Ano6 has no role in intestinal cholinergic Cl(-) secretion. Ano1 is located in a basolateral compartment/membrane rather than in the apical membrane, where it supports CCH-induced Ca(2+) increase, while the essential and possibly only apical Cl(-) channel is CFTR. These results define a new role of Ano1 for intestinal Ca(2+)-dependent Cl(-) secretion and demonstrate for the first time a contribution of Ano10 to intestinal transport.
Collapse
|
49
|
Hoffmann EK, Holm NB, Lambert IH. Functions of volume-sensitive and calcium-activated chloride channels. IUBMB Life 2014; 66:257-67. [PMID: 24771413 DOI: 10.1002/iub.1266] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2014] [Revised: 03/31/2014] [Accepted: 03/31/2014] [Indexed: 01/23/2023]
Abstract
The review describes molecular and functional properties of the volume regulated anion channel and Ca(2+)-dependent Cl(-) channels belonging to the anoctamin family with emphasis on physiological importance of these channels in regulation of cell volume, cell migration, cell proliferation, and programmed cell death. Finally, we discuss the role of Cl(-) channels in various diseases.
Collapse
Affiliation(s)
- Else Kay Hoffmann
- Department of Biology, University of Copenhagen, 13 Universitetsparken, Copenhagen Ø, Denmark
| | | | | |
Collapse
|
50
|
Zhang S, Chen Y, An H, Liu H, Li J, Pang C, Ji Q, Zhan Y. A novel biophysical model on calcium and voltage dual dependent gating of calcium-activated chloride channel. J Theor Biol 2014; 355:229-35. [PMID: 24727189 DOI: 10.1016/j.jtbi.2014.04.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2013] [Revised: 01/21/2014] [Accepted: 04/01/2014] [Indexed: 11/28/2022]
Abstract
Ca(2+)-activated Cl(-) channels (CaCCs) are anion-selective channels and involved in physiological processes such as electrolyte/fluid secretion, smooth muscle excitability, and olfactory perception which critically depend on the Ca(2+) and voltage dual-dependent gating of channels. However, how the Ca(2+) and voltage regulate the gating of CaCCs still unclear. In this work, the authors constructed a biophysical model to illustrate the dual-dependent gating of CaCCs. For validation, we applied our model on both native CaCCs and exogenous TMEM16A which is thought to be the molecular basis of CaCCs. Our data show that the native CaCCs may share universal gating mechanism. We confirmed the assumption that by binding with the channel, Ca(2+) decreases the energy-barrier to open the channel, but not changes the voltage-sensitivity. For TMEM16A, our model indicates that the exogenous channels show different Ca(2+) dependent gating mechanism from the native ones. These results advance the understanding of intracellular Ca(2+) and membrane potential regulation in CaCCs, and shed new light on its function in aspect of physiology and pharmacology.
Collapse
Affiliation(s)
- Suhua Zhang
- School of Sciences, Hebei University of Technology, Tianjin 300130, China
| | - Yafei Chen
- School of Sciences, Hebei University of Technology, Tianjin 300130, China
| | - Hailong An
- School of Sciences, Hebei University of Technology, Tianjin 300130, China
| | - Hui Liu
- School of Sciences, Hebei University of Technology, Tianjin 300130, China
| | - Junwei Li
- School of Sciences, Hebei University of Technology, Tianjin 300130, China
| | - Chunli Pang
- School of Sciences, Hebei University of Technology, Tianjin 300130, China
| | - Qing Ji
- School of Sciences, Hebei University of Technology, Tianjin 300130, China
| | - Yong Zhan
- School of Sciences, Hebei University of Technology, Tianjin 300130, China.
| |
Collapse
|