1
|
Motahari-Rad H, Subiri A, Soler R, Ocaña L, Alcaide J, Rodríguez-Capitan J, Buil V, el Azzouzi H, Ortega-Gomez A, Bernal-Lopez R, Insenser M, Tinahones FJ, Murri M. The Effect of Sex and Obesity on the Gene Expression of Lipid Flippases in Adipose Tissue. J Clin Med 2022; 11:jcm11133878. [PMID: 35807162 PMCID: PMC9267438 DOI: 10.3390/jcm11133878] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Revised: 06/25/2022] [Accepted: 06/28/2022] [Indexed: 01/25/2023] Open
Abstract
Molecular mechanisms behind obesity and sex-related effects in adipose tissue remain elusive. During adipocyte expansion, adipocytes undergo drastic remodelling of lipid membrane compositions. Lipid flippases catalyse phospholipid translocation from exoplasmic to the cytoplasmic leaflet of membranes. The present study aimed to analyse the effect of sex, obesity, and their interactions on the gene expression of two lipid flippases—ATP8A1 and ATP8B1—and their possible microRNA (miR) modulators in visceral adipose tissue (VAT). In total, 12 normal-weight subjects (5 premenopausal women and 7 men) and 13 morbidly obese patients (7 premenopausal women and 6 men) were submitted to surgery, and VAT samples were obtained. Gene expression levels of ATP8A1, ATP8B1, miR-548b-5p, and miR-4643 were measured in VAT. Our results showed a marked influence of obesity on VAT ATP8A1 and ATP8B1, although the effects of obesity were stronger in men for ATP8A1. Both genes positively correlated with obesity and metabolic markers. Furthermore, ATP8B1 was positively associated with miR-548b-5p and negatively associated with miR-4643. Both miRs were also affected by sex. Thus, lipid flippases are altered by obesity in VAT in a sex-specific manner. Our study provides a better understanding of the sex-specific molecular mechanisms underlying obesity, which may contribute to the development of sex-based precision medicine.
Collapse
Affiliation(s)
- Hanieh Motahari-Rad
- Department of Molecular Genetics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran 14117-13116, Iran;
- Clinical Management Unit (UGC) of Endocrinology and Nutrition, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Clínico Virgen de la Victoria, 29010 Málaga, Spain; (A.S.); (J.A.); (A.O.-G.); (F.J.T.)
| | - Alba Subiri
- Clinical Management Unit (UGC) of Endocrinology and Nutrition, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Clínico Virgen de la Victoria, 29010 Málaga, Spain; (A.S.); (J.A.); (A.O.-G.); (F.J.T.)
- CIBER Fisiopatología de la Obesidad y Nutrición (CIlBEROBN), Instituto de Salud Carlos III, 29010 Málaga, Spain;
| | - Rocio Soler
- Clinical Management Unit (UGC) of General and Digestive Surgery, Virgen de la Victoria University Hospital, 29010 Málaga, Spain; (R.S.); (L.O.)
| | - Luis Ocaña
- Clinical Management Unit (UGC) of General and Digestive Surgery, Virgen de la Victoria University Hospital, 29010 Málaga, Spain; (R.S.); (L.O.)
| | - Juan Alcaide
- Clinical Management Unit (UGC) of Endocrinology and Nutrition, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Clínico Virgen de la Victoria, 29010 Málaga, Spain; (A.S.); (J.A.); (A.O.-G.); (F.J.T.)
- CIBER Fisiopatología de la Obesidad y Nutrición (CIlBEROBN), Instituto de Salud Carlos III, 29010 Málaga, Spain;
| | - Jorge Rodríguez-Capitan
- Clinical Management Unit (UGC) of Heart, Virgen de la Victoria University Hospital, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Universitario Virgen de la Victoria, Universidad de Málaga (UMA), Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), 29010 Málaga, Spain;
- Faculty of Medicine, University of Malaga, 29010 Malaga, Spain;
| | - Veronica Buil
- Faculty of Medicine, University of Malaga, 29010 Malaga, Spain;
| | - Hamid el Azzouzi
- Department of Molecular Genetics, Erasmus University Medical Center, 3015 GD Rotterdam, The Netherlands;
| | - Almudena Ortega-Gomez
- Clinical Management Unit (UGC) of Endocrinology and Nutrition, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Clínico Virgen de la Victoria, 29010 Málaga, Spain; (A.S.); (J.A.); (A.O.-G.); (F.J.T.)
- CIBER Fisiopatología de la Obesidad y Nutrición (CIlBEROBN), Instituto de Salud Carlos III, 29010 Málaga, Spain;
| | - Rosa Bernal-Lopez
- CIBER Fisiopatología de la Obesidad y Nutrición (CIlBEROBN), Instituto de Salud Carlos III, 29010 Málaga, Spain;
- Clinical Management Unit (UGC) of Internal Medicine, IBIMA, Hospital Regional Universitario de Málaga, 29009 Málaga, Spain
| | - Maria Insenser
- Diabetes, Obesity and Human Reproduction Research Group, Department of Endocrinology & Nutrition, Hospital Universitario Ramón y Cajal & Universidad de Alcalá & Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS) & Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), 28034 Madrid, Spain
- Correspondence: (M.I.); (M.M.)
| | - Francisco J. Tinahones
- Clinical Management Unit (UGC) of Endocrinology and Nutrition, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Clínico Virgen de la Victoria, 29010 Málaga, Spain; (A.S.); (J.A.); (A.O.-G.); (F.J.T.)
- CIBER Fisiopatología de la Obesidad y Nutrición (CIlBEROBN), Instituto de Salud Carlos III, 29010 Málaga, Spain;
- Faculty of Medicine, University of Malaga, 29010 Malaga, Spain;
| | - Mora Murri
- Clinical Management Unit (UGC) of Endocrinology and Nutrition, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Clínico Virgen de la Victoria, 29010 Málaga, Spain; (A.S.); (J.A.); (A.O.-G.); (F.J.T.)
- CIBER Fisiopatología de la Obesidad y Nutrición (CIlBEROBN), Instituto de Salud Carlos III, 29010 Málaga, Spain;
- Correspondence: (M.I.); (M.M.)
| |
Collapse
|
2
|
Dietl P, Frick M. Channels and Transporters of the Pulmonary Lamellar Body in Health and Disease. Cells 2021; 11:45. [PMID: 35011607 PMCID: PMC8750383 DOI: 10.3390/cells11010045] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 12/20/2021] [Accepted: 12/22/2021] [Indexed: 02/06/2023] Open
Abstract
The lamellar body (LB) of the alveolar type II (ATII) cell is a lysosome-related organelle (LRO) that contains surfactant, a complex mix of mainly lipids and specific surfactant proteins. The major function of surfactant in the lung is the reduction of surface tension and stabilization of alveoli during respiration. Its lack or deficiency may cause various forms of respiratory distress syndrome (RDS). Surfactant is also part of the innate immune system in the lung, defending the organism against air-borne pathogens. The limiting (organelle) membrane that encloses the LB contains various transporters that are in part responsible for translocating lipids and other organic material into the LB. On the other hand, this membrane contains ion transporters and channels that maintain a specific internal ion composition including the acidic pH of about 5. Furthermore, P2X4 receptors, ligand gated ion channels of the danger signal ATP, are expressed in the limiting LB membrane. They play a role in boosting surfactant secretion and fluid clearance. In this review, we discuss the functions of these transporting pathways of the LB, including possible roles in disease and as therapeutic targets, including viral infections such as SARS-CoV-2.
Collapse
Affiliation(s)
- Paul Dietl
- Institute of General Physiology, Ulm University, Albert-Einstein-Allee 11, 89081 Ulm, Germany
| | - Manfred Frick
- Institute of General Physiology, Ulm University, Albert-Einstein-Allee 11, 89081 Ulm, Germany
| |
Collapse
|
3
|
Ko KW, Devault L, Sasaki Y, Milbrandt J, DiAntonio A. Live imaging reveals the cellular events downstream of SARM1 activation. eLife 2021; 10:e71148. [PMID: 34779400 PMCID: PMC8612704 DOI: 10.7554/elife.71148] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Accepted: 11/12/2021] [Indexed: 12/15/2022] Open
Abstract
SARM1 is an inducible NAD+ hydrolase that triggers axon loss and neuronal cell death in the injured and diseased nervous system. While SARM1 activation and enzyme function are well defined, the cellular events downstream of SARM1 activity but prior to axonal demise are much less well understood. Defects in calcium, mitochondria, ATP, and membrane homeostasis occur in injured axons, but the relationships among these events have been difficult to disentangle because prior studies analyzed large collections of axons in which cellular events occur asynchronously. Here, we used live imaging of mouse sensory neurons with single axon resolution to investigate the cellular events downstream of SARM1 activity. Our studies support a model in which SARM1 NADase activity leads to an ordered sequence of events from loss of cellular ATP, to defects in mitochondrial movement and depolarization, followed by calcium influx, externalization of phosphatidylserine, and loss of membrane permeability prior to catastrophic axonal self-destruction.
Collapse
Affiliation(s)
- Kwang Woo Ko
- Washington University School of MedicineSt LouisUnited States
| | - Laura Devault
- Washington University School of MedicineSt LouisUnited States
| | - Yo Sasaki
- Genetics, Washington University School of MedicineSt LouisUnited States
| | - Jeffrey Milbrandt
- Genetics, Hope Center for Neurological Disorders, Washington University School of MedicineSt LouisUnited States
| | - Aaron DiAntonio
- Developmental Biology, Needleman Center for Neurometabolism and Axonal Therapeutics, Washington University School of MedicineSt LouisUnited States
| |
Collapse
|
4
|
Davis HW, Kaynak A, Vallabhapurapu SD, Qi X. Targeting of elevated cell surface phosphatidylserine with saposin C-dioleoylphosphatidylserine nanodrug as individual or combination therapy for pancreatic cancer. World J Gastrointest Oncol 2021; 13:550-559. [PMID: 34163572 PMCID: PMC8204355 DOI: 10.4251/wjgo.v13.i6.550] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 04/13/2021] [Accepted: 05/10/2021] [Indexed: 02/06/2023] Open
Abstract
Pancreatic cancer is one of the deadliest of cancers with a five-year survival of roughly 8%. Current therapies are: surgery, radiation and chemotherapy. Surgery is curative only if the cancer is caught very early, which is rare, and the latter two modalities are only marginally effective and have significant side effects. We have developed a nanosome comprised of the lysosomal protein, saposin C (SapC) and the acidic phospholipid, dioleoylphosphatidylserine (DOPS). In the acidic tumor microenvironment, this molecule, SapC-DOPS, targets the phosphatidylserine cancer-biomarker which is predominantly elevated on the surface of cancer cells. Importantly, SapC-DOPS can selectively target pancreatic tumors and metastases. Furthermore, SapC-DOPS has exhibited an impressive safety profile with only a few minor side effects in both preclinical experiments and in phase I clinical trials. With the dismal outcomes for pancreatic cancer there is an urgent need for better treatments and SapC-DOPS is a good candidate for addition to the oncologist’s toolbox.
Collapse
Affiliation(s)
- Harold W Davis
- Division of Hematology/Oncology, Department of Internal Medicine, University of Cincinnati College of Medicine, Brain Tumor Center at UC Neuroscience Institute, Cincinnati, OH 45267, United States
| | - Ahmet Kaynak
- Division of Hematology/Oncology, Department of Internal Medicine, University of Cincinnati College of Medicine, Brain Tumor Center at UC Neuroscience Institute, Cincinnati, OH 45267, United States
- Department of Biomedical Engineering, College of Engineering and Applied Science, University of Cincinnati, Cincinnati, OH 45221, United States
| | - Subrahmanya D Vallabhapurapu
- Division of Hematology/Oncology, Department of Internal Medicine, University of Cincinnati College of Medicine, Brain Tumor Center at UC Neuroscience Institute, Cincinnati, OH 45267, United States
| | - Xiaoyang Qi
- Division of Hematology/Oncology, Department of Internal Medicine, University of Cincinnati College of Medicine, Brain Tumor Center at UC Neuroscience Institute, Cincinnati, OH 45267, United States
- Department of Biomedical Engineering, College of Engineering and Applied Science, University of Cincinnati, Cincinnati, OH 45221, United States
| |
Collapse
|
5
|
Yap YT, Li YH, Li W, Banerjee P, Zhang Z. ATP8a1, an IFT27 binding partner, is dispensable for spermatogenesis and male fertility. Mol Reprod Dev 2021; 88:371-375. [PMID: 33821543 DOI: 10.1002/mrd.23470] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 02/23/2021] [Indexed: 11/10/2022]
Abstract
Intraflagellar transport 27 (IFT27) is a key regulator for spermiogenesis and male fertility in mice. ATP8a1, a protein involved in the translocation of phosphatidylserine and phosphatidylethanolamine across lipid bilayers, is the strongest binding partner of IFT27. To investigate the role of ATP8a1 in spermatogenesis and male fertility, the global Atp8a1 knockout mice were analyzed. All mutant mice were fertile, and sperm count and motility were comparable to the control mice. Examination of testis and epididymis by hematoxylin and eosin staining did not reveal major histologic defects. These observations demonstrate that ATP8a1 is not a major spermatogenesis regulator. Given that a tissue-specific paralogue of ATP8a1, ATP8a2, is present, further studies with double-knockout models are warranted to delineate any compensatory functions of the two proteins.
Collapse
Affiliation(s)
- Yi T Yap
- Department of Physiology, Wayne State University, Detroit, Michigan, USA
| | - Yu H Li
- Department of Physiology, Wayne State University, Detroit, Michigan, USA.,School of Public Health, Wuhan University of Science and Technology, Wuhan, Hubei, China
| | - Wei Li
- Department of Physiology, Wayne State University, Detroit, Michigan, USA
| | - Probal Banerjee
- Department of Chemistry, The College of Staten Island (CUNY), Staten Island, New York, USA
| | - Zhibing Zhang
- Department of Physiology, Wayne State University, Detroit, Michigan, USA.,Department of Obstetrics & Gynecology, Wayne State University, Detroit, Michigan, USA
| |
Collapse
|
6
|
The transport mechanism of P4 ATPase lipid flippases. Biochem J 2021; 477:3769-3790. [PMID: 33045059 DOI: 10.1042/bcj20200249] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Revised: 09/02/2020] [Accepted: 09/16/2020] [Indexed: 12/18/2022]
Abstract
P4 ATPase lipid flippases are ATP-driven transporters that translocate specific lipids from the exoplasmic to the cytosolic leaflet of biological membranes, thus establishing a lipid gradient between the two leaflets that is essential for many cellular processes. While substrate specificity, subcellular and tissue-specific expression, and physiological functions have been assigned to a number of these transporters in several organisms, the mechanism of lipid transport has been a topic of intense debate in the field. The recent publication of a series of structural models based on X-ray crystallography and cryo-EM studies has provided the first glimpse into how P4 ATPases have adapted the transport mechanism used by the cation-pumping family members to accommodate a substrate that is at least an order of magnitude larger than cations.
Collapse
|
7
|
Bernecker C, Köfeler H, Pabst G, Trötzmüller M, Kolb D, Strohmayer K, Trajanoski S, Holzapfel GA, Schlenke P, Dorn I. Cholesterol Deficiency Causes Impaired Osmotic Stability of Cultured Red Blood Cells. Front Physiol 2019; 10:1529. [PMID: 31920725 PMCID: PMC6933518 DOI: 10.3389/fphys.2019.01529] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Accepted: 12/04/2019] [Indexed: 11/17/2022] Open
Abstract
Ex vivo generation of red blood cells (cRBCs) is an attractive tool in basic research and for replacing blood components donated by volunteers. As a prerequisite for the survival of cRBCs during storage as well as in the circulation, the quality of the membrane is of utmost importance. Besides the cytoskeleton and embedded proteins, the lipid bilayer is critical for membrane integrity. Although cRBCs suffer from increased fragility, studies investigating the lipid content of their membrane are still lacking. We investigated the membrane lipid profile of cRBCs from CD34+ human stem and progenitor cells compared to native red blood cells (nRBCs) and native reticulocytes (nRETs). Ex vivo erythropoiesis was performed in a well-established liquid assay. cRBCs showed a maturation grade between nRETs and nRBCs. High-resolution mass spectrometry analysis for cholesterol and the major phospholipid classes, phosphatidylcholine, phosphatidylethanolamine, phosphatidylinositol, phosphatidylserine, sphingomyelin and lysophosphatidylcholin, demonstrated severe cholesterol deficiency in cRBCs. Although cRBCs showed normal deformability capacity, they suffered from increased hemolysis due to minimal changes in the osmotic conditions. After additional lipid supplementation, especially cholesterol during culturing, the cholesterol content of cRBCs increased to a subnormal amount. Concurrently, the osmotic resistance recovered completely and became comparable to that of nRETs. Minor differences in the amount of phospholipids in cRBCs compared to native cells could mainly be attributed to the ongoing membrane remodeling process from the reticulocyte to the erythrocyte stage. Obtained results demonstrate severe cholesterol deficiency as a reason for enhanced fragility of cRBCs. Therefore, the supplementation of lipids, especially cholesterol during ex vivo erythropoiesis may overcome this limitation and strengthens the survival of cRBCs ex vivo and in vivo.
Collapse
Affiliation(s)
- Claudia Bernecker
- Department for Blood Group Serology and Transfusion Medicine, Medical University of Graz, Graz, Austria
| | - Harald Köfeler
- Center for Medical Research, Medical University of Graz, Graz, Austria
| | - Georg Pabst
- Institute of Molecular Biosciences, University of Graz, Biophysics Division, BioTechMed Graz, Graz, Austria
| | | | - Dagmar Kolb
- Center for Medical Research, Medical University of Graz, Graz, Austria.,Division of Cell Biology, Histology and Embryology, Gottfried Schatz Research Center, Medical University of Graz, Graz, Austria
| | - Karl Strohmayer
- Institute of Biomechanics, Graz University of Technology, Graz, Austria
| | - Slave Trajanoski
- Center for Medical Research, Medical University of Graz, Graz, Austria
| | - Gerhard A Holzapfel
- Institute of Biomechanics, Graz University of Technology, Graz, Austria.,Department of Structural Engineering, Norwegian University of Science and Technology, Trondheim, Norway
| | - Peter Schlenke
- Department for Blood Group Serology and Transfusion Medicine, Medical University of Graz, Graz, Austria
| | - Isabel Dorn
- Department for Blood Group Serology and Transfusion Medicine, Medical University of Graz, Graz, Austria
| |
Collapse
|
8
|
Liou AY, Molday LL, Wang J, Andersen JP, Molday RS. Identification and functional analyses of disease-associated P4-ATPase phospholipid flippase variants in red blood cells. J Biol Chem 2019; 294:6809-6821. [PMID: 30850395 DOI: 10.1074/jbc.ra118.007270] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Revised: 03/06/2019] [Indexed: 02/04/2023] Open
Abstract
ATP-dependent phospholipid flippase activity crucial for generating lipid asymmetry was first detected in red blood cell (RBC) membranes, but the P4-ATPases responsible have not been directly determined. Using affinity-based MS, we show that ATP11C is the only abundant P4-ATPase phospholipid flippase in human RBCs, whereas ATP11C and ATP8A1 are the major P4-ATPases in mouse RBCs. We also found that ATP11A and ATP11B are present at low levels. Mutations in the gene encoding ATP11C are responsible for blood and liver disorders, but the disease mechanisms are not known. Using heterologous expression, we show that the T415N substitution in the phosphorylation motif of ATP11C, responsible for congenital hemolytic anemia, reduces ATP11C expression, increases retention in the endoplasmic reticulum, and decreases ATPase activity by 61% relative to WT ATP11C. The I355K substitution in the transmembrane domain associated with cholestasis and anemia in mice was expressed at WT levels and trafficked to the plasma membrane but was devoid of activity. We conclude that the T415N variant causes significant protein misfolding, resulting in low protein expression, cellular mislocalization, and reduced functional activity. In contrast, the I355K variant folds and traffics normally but lacks key contacts required for activity. We propose that the loss in ATP11C phospholipid flippase activity coupled with phospholipid scramblase activity results in the exposure of phosphatidylserine on the surface of RBCs, decreasing RBC survival and resulting in anemia.
Collapse
Affiliation(s)
- Angela Y Liou
- From the Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, British Columbia V6T 1Z3, Canada and
| | - Laurie L Molday
- From the Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, British Columbia V6T 1Z3, Canada and
| | - Jiao Wang
- From the Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, British Columbia V6T 1Z3, Canada and
| | - Jens Peter Andersen
- Department of Biomedicine, Aarhus University, Ole Worms Allé 4, Building 1160, DK-8000 Aarhus C, Denmark
| | - Robert S Molday
- From the Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, British Columbia V6T 1Z3, Canada and
| |
Collapse
|
9
|
Davis HW, Vallabhapurapu SD, Chu Z, Vallabhapurapu SL, Franco RS, Mierzwa M, Kassing W, Barrett WL, Qi X. Enhanced phosphatidylserine-selective cancer therapy with irradiation and SapC-DOPS nanovesicles. Oncotarget 2019; 10:856-868. [PMID: 30783515 PMCID: PMC6368238 DOI: 10.18632/oncotarget.26615] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Accepted: 12/29/2018] [Indexed: 02/06/2023] Open
Abstract
Normal living cells exhibit phosphatidylserine (PS) primarily within the intracellular leaflet of the plasma membrane. In contrast, viable cancer cells have high levels of PS on the external surface, and exhibit a broad range of surface PS, even within specific types of cancer. Agents that target surface PS have recently been developed to treat tumors and are expected to be more effective with higher surface PS levels. In this context, we examined whether surface PS is increased with irradiation. In vitro irradiation of cancer cell lines selected surviving cells that had higher surface PS in a dose- and time-dependent manner. This was more pronounced if surface PS was initially in the lower range for cancer cells. Radiation also increased the surface PS of tumor cells in subcutaneous xenografts in nude mice. We found an inverse relationship between steady state surface PS level of cancer cell lines and their sensitivity to radiation-induced cell death. In addition, serial irradiation, which selected surviving cells with higher surface PS, also increased resistance to radiation and to some chemotherapeutic drugs, suggesting a PS-dependent mechanism for development of resistance to therapy. On the other hand, fractionated radiation enhanced the effect of a novel anti-cancer, PS-targeting drug, SapC-DOPS, in some cancer cell lines. Our data suggest that we can group cancer cells into cells with low surface PS, which are sensitive to radiation, and high surface PS, which are sensitive to SapC-DOPS. Combination of these interventions may provide a potential new combination therapy.
Collapse
Affiliation(s)
- Harold W Davis
- Division of Hematology/Oncology, Translational Research Laboratory, Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Subrahmanya D Vallabhapurapu
- Division of Hematology/Oncology, Translational Research Laboratory, Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Zhengtao Chu
- Division of Hematology/Oncology, Translational Research Laboratory, Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Swarajya L Vallabhapurapu
- Division of Hematology/Oncology, Translational Research Laboratory, Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Robert S Franco
- Division of Hematology/Oncology, Translational Research Laboratory, Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Michelle Mierzwa
- Department of Radiation Oncology, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - William Kassing
- Department of Radiation Oncology, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - William L Barrett
- Department of Radiation Oncology, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Xiaoyang Qi
- Division of Hematology/Oncology, Translational Research Laboratory, Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, USA.,Division of Human Genetics, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| |
Collapse
|
10
|
Li Q, Yang Y, Liu Y. Over-Expression of ATPase II Alleviates Ethanol-Induced Hepatocyte Injury in HL-7702 Cells. Med Sci Monit 2018; 24:8372-8382. [PMID: 30457983 PMCID: PMC6256429 DOI: 10.12659/msm.910254] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Background Excessive alcohol consumption can cause hepatocellular injury. ATPase II (ATP8A1) can display an ATP-dependent phospholipid translocase activity. However, the function of ATP8A1 in hepatocyte injury is still unclear. In the present study we explored the effect of ATP8A1 on ethanol-induced hepatocyte injury. Material/Method A human hepatocyte strain, HL-7702, was pretreated by ethanol with gradient concentration for 2, 4, 8, and 12 h, and were then divided into 6 groups after the cells were transfected. We detected cell viability by use of the Cell Counting Kit-8 (CCK-8) assay. Reactive oxygen species (ROS), apoptosis rate, and mitochondrial membrane potential (MMP) were measured using flow cytometry. We used quantitative reverse transcription-polymerase chain reaction (qRT-PCR) and Western blot to measure the mRNA and protein expression, respectively. Results Ethanol inhibited the viability of HL-7702 cells and suppressed the expression of ATP8A1 in dose- and time-dependent manners. Furthermore, over-expression of ATP8A1 reduced the level of ROS and the apoptosis rate and recovered the MMP. Additionally, over-expressed ATP8A1 regulated the protein and mRNA levels of apoptosis-related molecules. Moreover, over-expression of ATP8A1 enhanced the phosphorylation of phosphatidylinositol 3-kinase (PI3K) and protein kinase B (Akt). Conclusions Over-expression of ATP8A1 alleviated ethanol-induced hepatocyte injury. Moreover, the PI3K/Akt signaling pathway appears to participate in inhibition of ethanol-induced hepatocyte apoptosis and may provide a candidate target for the treatment of alcoholic liver diseases (ALD).
Collapse
Affiliation(s)
- Qing Li
- Department of Clinical Laboratory, Jingmen No. 1 People's Hospital, Jingmen, Hubei, China (mainland)
| | - Yan Yang
- Department of Clinical Laboratory, Jingmen No. 1 People's Hospital, Jingmen, Hubei, China (mainland)
| | - Ying Liu
- Department of Clinical Laboratory, Jingmen No. 1 People's Hospital, Jingmen, Hubei, China (mainland)
| |
Collapse
|
11
|
Cotton RJ, Ploier B, Goren MA, Menon AK, Graumann J. flippant-An R package for the automated analysis of fluorescence-based scramblase assays. BMC Bioinformatics 2017; 18:146. [PMID: 28253836 PMCID: PMC5335725 DOI: 10.1186/s12859-017-1542-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2016] [Accepted: 02/08/2017] [Indexed: 11/30/2022] Open
Abstract
Background The lipid scrambling activity of protein extracts and purified scramblases is typically measured using a fluorescence-based assay. While the assay has yielded insight into the scramblase activity in crude membrane preparations, functional validation of candidate scramblases, stoichiometry of scramblase complexes as well as ATP-dependence of flippases, data analysis in its context has remained a task involving many manual steps. Results With the extension package “flippant” to R, a free software environment for statistical computing and graphics, we introduce an integrated solution for the analysis and publication-grade graphical presentation of dithionite scramblase assays and demonstrate its utility in revisiting an originally manual analysis from the publication record, closely reproducing the reported results. Conclusions “flippant” allows for quick, reproducible data analysis of scramblase activity assays and provides a platform for review, dissemination and extension of the strategies it employs. Electronic supplementary material The online version of this article (doi:10.1186/s12859-017-1542-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Richard J Cotton
- Research Division, Weill Cornell Medicine-Qatar, P.O.Box 24144, Doha, State of Qatar
| | - Birgit Ploier
- Department of Biochemistry, Weill Cornell Medical College, New York, NY, 10065, USA
| | - Michael A Goren
- Department of Biochemistry, Weill Cornell Medical College, New York, NY, 10065, USA
| | - Anant K Menon
- Department of Biochemistry, Weill Cornell Medical College, New York, NY, 10065, USA
| | - Johannes Graumann
- Research Division, Weill Cornell Medicine-Qatar, P.O.Box 24144, Doha, State of Qatar. .,Current Address: Scientific Service Group Biomolecular Mass Spectrometry, Max Planck Institute for Heart and Lung Research, W.G. Kerckhoff Institute, Ludwigstr. 43, D-61231, Bad Nauheim, Germany.
| |
Collapse
|
12
|
Vallabhapurapu SD, Blanco VM, Sulaiman MK, Vallabhapurapu SL, Chu Z, Franco RS, Qi X. Variation in human cancer cell external phosphatidylserine is regulated by flippase activity and intracellular calcium. Oncotarget 2016; 6:34375-88. [PMID: 26462157 PMCID: PMC4741459 DOI: 10.18632/oncotarget.6045] [Citation(s) in RCA: 91] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2015] [Accepted: 09/09/2015] [Indexed: 01/05/2023] Open
Abstract
Viable cancer cells expose elevated levels of phosphatidylserine (PS) on the exoplasmic face of the plasma membrane. However, the mechanisms leading to elevated PS exposure in viable cancer cells have not been defined. We previously showed that externalized PS may be used to monitor, target and kill tumor cells. In addition, PS on tumor cells is recognized by macrophages and has implications in antitumor immunity. Therefore, it is important to understand the molecular details of PS exposure on cancer cells in order to improve therapeutic targeting. Here we explored the mechanisms regulating the surface PS exposure in human cancer cells and found that differential flippase activity and intracellular calcium are the major regulators of surface PS exposure in viable human cancer cells. In general, cancer cell lines with high surface PS exhibited low flippase activity and high intracellular calcium, whereas cancer cells with low surface PS exhibited high flippase activity and low intracellular calcium. High surface PS cancer cells also had higher total cellular PS than low surface PS cells. Together, our results indicate that the amount of external PS in cancer cells is regulated by calcium dependent flippase activity and may also be influenced by total cellular PS.
Collapse
Affiliation(s)
- Subrahmanya D Vallabhapurapu
- Division of Hematology and Oncology, Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Víctor M Blanco
- Division of Hematology and Oncology, Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Mahaboob K Sulaiman
- Division of Hematology and Oncology, Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Swarajya Lakshmi Vallabhapurapu
- Division of Hematology and Oncology, Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Zhengtao Chu
- Division of Hematology and Oncology, Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA.,Divison of Human Genetics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Robert S Franco
- Division of Hematology and Oncology, Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Xiaoyang Qi
- Division of Hematology and Oncology, Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA.,Divison of Human Genetics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| |
Collapse
|
13
|
Pomorski TG, Menon AK. Lipid somersaults: Uncovering the mechanisms of protein-mediated lipid flipping. Prog Lipid Res 2016; 64:69-84. [PMID: 27528189 DOI: 10.1016/j.plipres.2016.08.003] [Citation(s) in RCA: 128] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2016] [Accepted: 08/10/2016] [Indexed: 12/22/2022]
Abstract
Membrane lipids diffuse rapidly in the plane of the membrane but their ability to flip spontaneously across a membrane bilayer is hampered by a significant energy barrier. Thus spontaneous flip-flop of polar lipids across membranes is very slow, even though it must occur rapidly to support diverse aspects of cellular life. Here we discuss the mechanisms by which rapid flip-flop occurs, and what role lipid flipping plays in membrane homeostasis and cell growth. We focus on conceptual aspects, highlighting mechanistic insights from biochemical and in silico experiments, and the recent, ground-breaking identification of a number of lipid scramblases.
Collapse
Affiliation(s)
- Thomas Günther Pomorski
- Faculty of Chemistry and Biochemistry, Molecular Biochemistry, Ruhr University Bochum, Universitätstrasse 150, D-44780 Bochum, Germany; Centre for Membrane Pumps in Cells and Disease-PUMPKIN, Department of Plant and Environmental Sciences, University of Copenhagen, Frederiksberg C, Denmark.
| | - Anant K Menon
- Department of Biochemistry, Weill Cornell Medical College, 1300 York Avenue, New York, NY 10065, USA.
| |
Collapse
|
14
|
Chaubey PM, Hofstetter L, Roschitzki B, Stieger B. Proteomic Analysis of the Rat Canalicular Membrane Reveals Expression of a Complex System of P4-ATPases in Liver. PLoS One 2016; 11:e0158033. [PMID: 27347675 PMCID: PMC4922570 DOI: 10.1371/journal.pone.0158033] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2015] [Accepted: 06/09/2016] [Indexed: 12/22/2022] Open
Abstract
Transport processes in the canalicular membrane are key elements in bile formation and are the driving force of the enterohepatic circulation of bile salts. The canalicular membrane is constantly exposed to the detergent action of bile salts. One potential element protecting the canalicular membrane from the high canalicular bile salt concentrations may be bile salt resistant microdomains, however additional factors are likely to play a role. To obtain more insights into the molecular composition of the canalicular membrane, the proteome of highly purified rat canalicular membrane vesicles was determined. Isolated rat canalicular membrane vesicles were stripped from adhering proteins, deglycosylated and protease digested before subjecting the samples to shot gun proteomic analysis. The expression of individual candidates was studied by PCR, Western blotting and immunohistochemistry. A total of 2449 proteins were identified, of which 1282 were predicted to be membrane proteins. About 50% of the proteins identified here were absent from previously published liver proteomes. In addition to ATP8B1, four more P4-ATPases were identified. ATP8A1 and ATP9A showed expression specific to the canalicular membrane, ATP11C at the bLPM and ATP11A in an intracellular vesicular compartment partially colocalizing with RAB7A and EEA1 as markers of the endosomal compartment. This study helped to identify additional P4-ATPases from rat liver particularly in the canalicular membrane, previously not known to be expressed in liver. These P4-ATPases might be contributing for maintaining transmembrane lipid homeostasis in hepatocytes.
Collapse
Affiliation(s)
- Pururawa Mayank Chaubey
- Department of Clinical Pharmacology and Toxicology, University Hospital Zürich, Zürich, Switzerland
| | - Lia Hofstetter
- Department of Clinical Pharmacology and Toxicology, University Hospital Zürich, Zürich, Switzerland
| | - Bernd Roschitzki
- Functional Genomics Center Zürich, University of Zürich/ETH Zürich, Zürich, Switzerland
| | - Bruno Stieger
- Department of Clinical Pharmacology and Toxicology, University Hospital Zürich, Zürich, Switzerland
- * E-mail:
| |
Collapse
|
15
|
Andersen JP, Vestergaard AL, Mikkelsen SA, Mogensen LS, Chalat M, Molday RS. P4-ATPases as Phospholipid Flippases-Structure, Function, and Enigmas. Front Physiol 2016; 7:275. [PMID: 27458383 PMCID: PMC4937031 DOI: 10.3389/fphys.2016.00275] [Citation(s) in RCA: 234] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2016] [Accepted: 06/20/2016] [Indexed: 01/26/2023] Open
Abstract
P4-ATPases comprise a family of P-type ATPases that actively transport or flip phospholipids across cell membranes. This generates and maintains membrane lipid asymmetry, a property essential for a wide variety of cellular processes such as vesicle budding and trafficking, cell signaling, blood coagulation, apoptosis, bile and cholesterol homeostasis, and neuronal cell survival. Some P4-ATPases transport phosphatidylserine and phosphatidylethanolamine across the plasma membrane or intracellular membranes whereas other P4-ATPases are specific for phosphatidylcholine. The importance of P4-ATPases is highlighted by the finding that genetic defects in two P4-ATPases ATP8A2 and ATP8B1 are associated with severe human disorders. Recent studies have provided insight into how P4-ATPases translocate phospholipids across membranes. P4-ATPases form a phosphorylated intermediate at the aspartate of the P-type ATPase signature sequence, and dephosphorylation is activated by the lipid substrate being flipped from the exoplasmic to the cytoplasmic leaflet similar to the activation of dephosphorylation of Na(+)/K(+)-ATPase by exoplasmic K(+). How the phospholipid is translocated can be understood in terms of a peripheral hydrophobic gate pathway between transmembrane helices M1, M3, M4, and M6. This pathway, which partially overlaps with the suggested pathway for migration of Ca(2+) in the opposite direction in the Ca(2+)-ATPase, is wider than the latter, thereby accommodating the phospholipid head group. The head group is propelled along against its concentration gradient with the hydrocarbon chains projecting out into the lipid phase by movement of an isoleucine located at the position corresponding to an ion binding glutamate in the Ca(2+)- and Na(+)/K(+)-ATPases. Hence, the P4-ATPase mechanism is quite similar to the mechanism of these ion pumps, where the glutamate translocates the ions by moving like a pump rod. The accessory subunit CDC50 may be located in close association with the exoplasmic entrance of the suggested pathway, and possibly promotes the binding of the lipid substrate. This review focuses on properties of mammalian and yeast P4-ATPases for which most mechanistic insight is available. However, the structure, function and enigmas associated with mammalian and yeast P4-ATPases most likely extend to P4-ATPases of plants and other organisms.
Collapse
Affiliation(s)
| | | | | | | | - Madhavan Chalat
- Department of Biochemistry and Molecular Biology, University of British ColumbiaVancouver, BC, Canada
| | - Robert S. Molday
- Department of Biochemistry and Molecular Biology, University of British ColumbiaVancouver, BC, Canada
- *Correspondence: Robert S. Molday
| |
Collapse
|
16
|
Dong W, Yao C, Teng X, Chai J, Yang X, Li B. MiR-140-3p suppressed cell growth and invasion by downregulating the expression of ATP8A1 in non-small cell lung cancer. Tumour Biol 2015; 37:2973-85. [PMID: 26415732 DOI: 10.1007/s13277-015-3452-9] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2015] [Accepted: 04/10/2015] [Indexed: 12/21/2022] Open
Abstract
MicroRNAs (miRNAs) as a class of small noncoding RNA molecules regulate the expression of targeted gene. The dysregulation of microRNAs is reported to be involved in carcinogenesis and tumor progression. Here, we identified miR-140-3p as a downregulated microRNA in most cancer tissues including lung cancer tissues, compared with their normal counterparts. MiR-140-3p was observed to perform its tumor suppressor function via its inhibition on cell growth, migration and invasion but its induction of cell apoptosis. Furthermore, the growth of non-small-cell lung cancer (NSCLC) cells in nude mouse models were suppressed by overexpression of miR-140-3p. ATP8A1 was demonstrated as a novel direct target of miR-140-3p using a luciferase assay. The increased level of intracellular ATP8A1 protein attenuated the inhibitor role of miR-140-3p in the growth and mobility of NSCLC cell. A regulation mechanism of miR-140-3p for the development and progression of NSCLC through downregulating the ATP8A1 expression was first discovered in the present study.
Collapse
Affiliation(s)
- Wei Dong
- Shandong University School of Medicine, 44# Wenhua Xi Road, Jinan, 250012, Shandong, People's Republic of China
| | - Chunping Yao
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, 440# Jiyan Road, Jinan, 250117, Shandong, People's Republic of China
| | - Xuepeng Teng
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, 440# Jiyan Road, Jinan, 250117, Shandong, People's Republic of China
| | - Jie Chai
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, 440# Jiyan Road, Jinan, 250117, Shandong, People's Republic of China
| | - Xinhua Yang
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, 440# Jiyan Road, Jinan, 250117, Shandong, People's Republic of China
| | - Baosheng Li
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, 440# Jiyan Road, Jinan, 250117, Shandong, People's Republic of China.
| |
Collapse
|
17
|
P4-ATPases: lipid flippases in cell membranes. Pflugers Arch 2015; 466:1227-40. [PMID: 24077738 PMCID: PMC4062807 DOI: 10.1007/s00424-013-1363-4] [Citation(s) in RCA: 95] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2013] [Revised: 09/11/2013] [Accepted: 09/11/2013] [Indexed: 12/13/2022]
Abstract
Cellular membranes, notably eukaryotic plasma membranes, are equipped with special proteins that actively translocate lipids from one leaflet to the other and thereby help generate membrane lipid asymmetry. Among these ATP-driven transporters, the P4 subfamily of P-type ATPases (P4-ATPases) comprises lipid flippases that catalyze the translocation of phospholipids from the exoplasmic to the cytosolic leaflet of cell membranes. While initially characterized as aminophospholipid translocases, recent studies of individual P4-ATPase family members from fungi, plants, and animals show that P4-ATPases differ in their substrate specificities and mediate transport of a broader range of lipid substrates, including lysophospholipids and synthetic alkylphospholipids. At the same time, the cellular processes known to be directly or indirectly affected by this class of transporters have expanded to include the regulation of membrane traffic, cytoskeletal dynamics, cell division, lipid metabolism, and lipid signaling. In this review, we will summarize the basic features of P4-ATPases and the physiological implications of their lipid transport activity in the cell.
Collapse
|
18
|
Lee S, Uchida Y, Wang J, Matsudaira T, Nakagawa T, Kishimoto T, Mukai K, Inaba T, Kobayashi T, Molday RS, Taguchi T, Arai H. Transport through recycling endosomes requires EHD1 recruitment by a phosphatidylserine translocase. EMBO J 2015; 34:669-88. [PMID: 25595798 PMCID: PMC4365035 DOI: 10.15252/embj.201489703] [Citation(s) in RCA: 110] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
P4-ATPases translocate aminophospholipids, such as phosphatidylserine (PS), to the cytosolic leaflet of membranes. PS is highly enriched in recycling endosomes (REs) and is essential for endosomal membrane traffic. Here, we show that PS flipping by an RE-localized P4-ATPase is required for the recruitment of the membrane fission protein EHD1. Depletion of ATP8A1 impaired the asymmetric transbilayer distribution of PS in REs, dissociated EHD1 from REs, and generated aberrant endosomal tubules that appear resistant to fission. EHD1 did not show membrane localization in cells defective in PS synthesis. ATP8A2, a tissue-specific ATP8A1 paralogue, is associated with a neurodegenerative disease (CAMRQ). ATP8A2, but not the disease-causative ATP8A2 mutant, rescued the endosomal defects in ATP8A1-depleted cells. Primary neurons from Atp8a2-/- mice showed a reduced level of transferrin receptors at the cell surface compared to Atp8a2+/+ mice. These findings demonstrate the role of P4-ATPase in membrane fission and give insight into the molecular basis of CAMRQ.
Collapse
Affiliation(s)
- Shoken Lee
- Department of Health Chemistry, Graduate School of Pharmaceutical Sciences University of Tokyo, Tokyo, Japan
| | - Yasunori Uchida
- Department of Health Chemistry, Graduate School of Pharmaceutical Sciences University of Tokyo, Tokyo, Japan
| | - Jiao Wang
- Departments of Biochemistry and Molecular Biology and Ophthalmology and Visual Sciences, Centre for Macular Research University of British Columbia, Vancouver, BC, Canada
| | - Tatsuyuki Matsudaira
- Department of Health Chemistry, Graduate School of Pharmaceutical Sciences University of Tokyo, Tokyo, Japan
| | - Takatoshi Nakagawa
- Department of Pharmacology, Osaka Medical College, Takatsuki-city Osaka, Japan
| | | | - Kojiro Mukai
- Department of Health Chemistry, Graduate School of Pharmaceutical Sciences University of Tokyo, Tokyo, Japan Lipid Biology Laboratory, RIKEN, Wako-shi Saitama, Japan
| | - Takehiko Inaba
- Lipid Biology Laboratory, RIKEN, Wako-shi Saitama, Japan
| | | | - Robert S Molday
- Departments of Biochemistry and Molecular Biology and Ophthalmology and Visual Sciences, Centre for Macular Research University of British Columbia, Vancouver, BC, Canada
| | - Tomohiko Taguchi
- Department of Health Chemistry, Graduate School of Pharmaceutical Sciences University of Tokyo, Tokyo, Japan Pathological Cell Biology Laboratory, Graduate School of Pharmaceutical Sciences University of Tokyo, Tokyo, Japan
| | - Hiroyuki Arai
- Department of Health Chemistry, Graduate School of Pharmaceutical Sciences University of Tokyo, Tokyo, Japan Pathological Cell Biology Laboratory, Graduate School of Pharmaceutical Sciences University of Tokyo, Tokyo, Japan
| |
Collapse
|
19
|
Gulshan K, Smith J. Sphingomyelin regulation of plasma membrane asymmetry, efflux and reverse cholesterol transport. ACTA ACUST UNITED AC 2014. [DOI: 10.2217/clp.14.28] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
|
20
|
Structure and mechanism of ATP-dependent phospholipid transporters. Biochim Biophys Acta Gen Subj 2014; 1850:461-75. [PMID: 24746984 DOI: 10.1016/j.bbagen.2014.04.008] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2014] [Revised: 04/04/2014] [Accepted: 04/07/2014] [Indexed: 01/09/2023]
Abstract
BACKGROUND ATP-binding cassette (ABC) transporters and P4-ATPases are two large and seemingly unrelated families of primary active pumps involved in moving phospholipids from one leaflet of a biological membrane to the other. SCOPE OF REVIEW This review aims to identify common mechanistic features in the way phospholipid flipping is carried out by two evolutionarily unrelated families of transporters. MAJOR CONCLUSIONS Both protein families hydrolyze ATP, although they employ different mechanisms to use it, and have a comparable size with twelve transmembrane segments in the functional unit. Further, despite differences in overall architecture, both appear to operate by an alternating access mechanism and during transport they might allow access of phospholipids to the internal part of the transmembrane domain. The latter feature is obvious for ABC transporters, but phospholipids and other hydrophobic molecules have also been found embedded in P-type ATPase crystal structures. Taken together, in two diverse groups of pumps, nature appears to have evolved quite similar ways of flipping phospholipids. GENERAL SIGNIFICANCE Our understanding of the structural basis for phospholipid flipping is still limited but it seems plausible that a general mechanism for phospholipid flipping exists in nature. This article is part of a Special Issue entitled Structural biochemistry and biophysics of membrane proteins.
Collapse
|
21
|
Kuypers FA. Hemoglobin S Polymerization and Red Cell Membrane Changes. Hematol Oncol Clin North Am 2014; 28:155-79. [DOI: 10.1016/j.hoc.2013.12.002] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
|
22
|
van der Mark VA, Elferink RPJO, Paulusma CC. P4 ATPases: flippases in health and disease. Int J Mol Sci 2013; 14:7897-922. [PMID: 23579954 PMCID: PMC3645723 DOI: 10.3390/ijms14047897] [Citation(s) in RCA: 101] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2013] [Revised: 03/28/2013] [Accepted: 04/07/2013] [Indexed: 12/26/2022] Open
Abstract
P4 ATPases catalyze the translocation of phospholipids from the exoplasmic to the cytosolic leaflet of biological membranes, a process termed “lipid flipping”. Accumulating evidence obtained in lower eukaryotes points to an important role for P4 ATPases in vesicular protein trafficking. The human genome encodes fourteen P4 ATPases (fifteen in mouse) of which the cellular and physiological functions are slowly emerging. Thus far, deficiencies of at least two P4 ATPases, ATP8B1 and ATP8A2, are the cause of severe human disease. However, various mouse models and in vitro studies are contributing to our understanding of the cellular and physiological functions of P4-ATPases. This review summarizes current knowledge on the basic function of these phospholipid translocating proteins, their proposed action in intracellular vesicle transport and their physiological role.
Collapse
Affiliation(s)
- Vincent A van der Mark
- Tytgat Institute for Liver and Intestinal Research, Academic Medical Center, Meibergdreef 69-71, 1105 BK Amsterdam, The Netherlands.
| | | | | |
Collapse
|
23
|
Levano K, Punia V, Raghunath M, Debata PR, Curcio GM, Mogha A, Purkayastha S, McCloskey D, Fata J, Banerjee P. Atp8a1 deficiency is associated with phosphatidylserine externalization in hippocampus and delayed hippocampus-dependent learning. J Neurochem 2011; 120:302-13. [PMID: 22007859 DOI: 10.1111/j.1471-4159.2011.07543.x] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The molecule responsible for the enzyme activity plasma membrane (PM) aminophospholipid translocase (APLT), which catalyzes phosphatidylserine (PS) translocation from the outer to the inner leaflet of the plasma membrane, is unknown in mammals. A Caenorhabditis elegans study has shown that ablation of transbilayer amphipath transporter-1 (TAT-1), which is an ortholog of a mammalian P-type ATPase, Atp8a1, causes PS externalization in the germ cells. We demonstrate here that the hippocampal cells of the dentate gyrus, and Cornu Ammonis (CA1, CA3) in mice lacking Atp8a1 exhibit a dramatic increase in PS externalization. Although their hippocampi showed no abnormal morphology or heightened apoptosis, these mice displayed increased activity and a marked deficiency in hippocampus-dependent learning, but no hyper-anxiety. Such observations indicate that Atp8a1 plays a crucial role in PM-APLT activity in the neuronal cells. In corroboration, ectopic expression of Atp8a1 but not its close homolog, Atp8a2, caused an increase in the population (V(max) ) of PM-APLT without any change in its signature parameter K(m) in the neuronal N18 cells. Conversely, expression of a P-type phosphorylation-site mutant of Atp8a1 (Atp8a1*) caused a decrease in V(max) of PM-APLT without significantly altering its K(m) . The Atp8a1*-expressing N18 cells also exhibited PS externalization without apoptosis. Together, our data strongly indicate that Atp8a1 plays a central role in the PM-APLT activity of some mammalian cells, such as the neuronal N18 and hippocampal cells.
Collapse
Affiliation(s)
- Kelly Levano
- CUNY Doctoral Program in Biochemistry, City University of New York at the College of Staten Island, Staten Island, New York, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Sharom FJ. Flipping and flopping--lipids on the move. IUBMB Life 2011; 63:736-46. [PMID: 21793163 DOI: 10.1002/iub.515] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2011] [Accepted: 05/16/2011] [Indexed: 12/24/2022]
Abstract
The rapid movement of polar lipids from one membrane leaflet to the other is facilitated by lipid flippases or translocases. Although their activity was first observed over 30 years ago, the structures, physiological roles, and molecular mechanisms of this group of proteins remain enigmatic. Lipid flippases maintain membrane lipid asymmetry, and in eukaryotes they are also intimately involved in membrane budding and vesicle trafficking. The ATP-dependent flippases are members of well-characterized protein families, whose other members transport nonlipid substrates across cell membranes. The P(4)-type ATPases carry out the inward translocation of phospholipids, and various ABC transporters are involved in outward lipid movement. The ATP-independent flippases move lipid substrates in both directions between membrane leaflets. With only a few exceptions, the molecular identity of these proteins is still unknown, despite their involvement in key biosynthetic pathways in both bacteria and eukaryotes. This review provides an overview of the different classes of flippases, and summarizes recent progress in their identification and functional characterization. The possible mechanisms of action of lipid flippases are discussed, and future directions explored.
Collapse
Affiliation(s)
- Frances J Sharom
- Department of Molecular and Cellular Biology, Biophysics Interdepartmental Group, University of Guelph, Guelph, ON, Canada.
| |
Collapse
|