Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 128
Filtrar
1.
Nat Plants ; 7(9): 1188-1199, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34531559

RESUMO

Lipid flippases (P4 ATPases) are active transporters that catalyse the translocation of lipids between the two sides of the biological membranes in the secretory pathway. This activity modulates biological membrane properties, contributes to vesicle formation, and is the trigger for lipid signalling events, which makes P4 ATPases essential for eukaryotic cell survival. Plant P4 ATPases (also known as aminophospholipid ATPases (ALAs)) are crucial for plant fertility and proper development, and are involved in key adaptive responses to biotic and abiotic stress, including chilling tolerance, heat adaptation, nutrient deficiency responses and pathogen defence. While ALAs present many analogies to mammalian and yeast P4 ATPases, they also show characteristic features as the result of their independent evolution. In this Review, the main properties, roles, regulation and mechanisms of action of ALA proteins are discussed.


Assuntos
Adaptação Fisiológica/fisiologia , Metabolismo dos Lipídeos/fisiologia , Proteínas de Transferência de Fosfolipídeos/fisiologia , Fenômenos Fisiológicos Vegetais , Estresse Fisiológico/fisiologia
2.
J Virol ; 95(20): e0116521, 2021 09 27.
Artigo em Inglês | MEDLINE | ID: mdl-34319156

RESUMO

Ebola virus (EBOV) attaches to target cells using two categories of cell surface receptors: C-type lectins and phosphatidylserine (PS) receptors. PS receptors typically bind to apoptotic cell membrane PS and orchestrate the uptake and clearance of apoptotic debris. Many enveloped viruses also contain exposed PS and can therefore exploit these receptors for cell entry. Viral infection can induce PS externalization in host cells, resulting in increased outer PS levels on budding virions. Scramblase enzymes carry out cellular PS externalization; thus, we targeted these proteins in order to manipulate viral envelope PS levels. We investigated two scramblases previously identified to be involved in EBOV PS levels, transmembrane protein 16F and Xk-related protein 8 (XKR8), as possible mediators of cellular and viral envelope surface PS levels during the replication of recombinant vesicular stomatitis virus containing its native glycoprotein (rVSV/G) or the EBOV glycoprotein (rVSV/EBOV-GP). We found that rVSV/G and rVSV/EBOV-GP virions produced in XKR8 knockout cells contain decreased levels of PS on their surfaces, and the PS-deficient rVSV/EBOV-GP virions are 70% less efficient at infecting cells through PS receptors. We also observed reduced rVSV and EBOV virus-like particle (VLP) budding in ΔXKR8 cells. Deletion of XKR8 in HAP1 cells reduced rVSV/G and rVSV/EBOV-GP budding by 60 and 65%, respectively, and reduced Ebola VLP budding more than 60%. We further demonstrated that caspase cleavage of XKR8 is required to promote budding. This suggests that XKR8, in addition to mediating virion PS levels, may also be critical for enveloped virus budding at the plasma membrane. IMPORTANCE Within the last decade, countries in western and central Africa have experienced the most widespread and deadly Ebola outbreaks since Ebola virus was identified in 1976. While outbreaks are primarily attributed to zoonotic transfer events, new evidence is emerging outbreaks may be caused by a combination of spillover events and viral latency or persistence in survivors. The possibility that Ebola virus can remain dormant and then reemerge in survivors highlights the critical need to prevent the virus from entering and establishing infection in human cells. Thus far, host cell scramblases TMEM16F and XKR8 have been implicated in Ebola envelope surface phosphatidylserine (PS) and cell entry using PS receptors. We assessed the contributions of these proteins using CRISPR knockout cells and two EBOV models: rVSV/EBOV-GP and EBOV VLPs. We observed that XKR8 is required for optimal EBOV envelope PS levels and infectivity and particle budding across all viral models.


Assuntos
Ebolavirus/metabolismo , Fosfatidilserinas/metabolismo , Liberação de Vírus/fisiologia , Linhagem Celular , Ebolavirus/patogenicidade , Glicoproteínas/metabolismo , Doença pelo Vírus Ebola/virologia , Humanos , Fosfatidilserinas/fisiologia , Proteínas de Transferência de Fosfolipídeos/metabolismo , Proteínas de Transferência de Fosfolipídeos/fisiologia , Proteínas do Envelope Viral/metabolismo , Vírion/metabolismo , Montagem de Vírus/genética , Montagem de Vírus/fisiologia , Liberação de Vírus/genética
3.
Proc Natl Acad Sci U S A ; 118(16)2021 04 20.
Artigo em Inglês | MEDLINE | ID: mdl-33850023

RESUMO

The autophagy protein ATG2, proposed to transfer bulk lipid from the endoplasmic reticulum (ER) during autophagosome biogenesis, interacts with ER residents TMEM41B and VMP1 and with ATG9, in Golgi-derived vesicles that initiate autophagosome formation. In vitro assays reveal TMEM41B, VMP1, and ATG9 as scramblases. We propose a model wherein membrane expansion results from the partnership of a lipid transfer protein, moving lipids between the cytosolic leaflets of apposed organelles, and scramblases that reequilibrate the leaflets of donor and acceptor organelle membranes as lipids are depleted or augmented. TMEM41B and VMP1 are implicated broadly in lipid homeostasis and membrane dynamics processes in which their scrambling activities likely are key.


Assuntos
Proteínas Relacionadas à Autofagia/metabolismo , Proteínas de Transferência de Fosfolipídeos/metabolismo , Autofagossomos/metabolismo , Autofagia/fisiologia , Proteínas Relacionadas à Autofagia/fisiologia , Proteínas de Transporte/metabolismo , Retículo Endoplasmático/metabolismo , Humanos , Metabolismo dos Lipídeos/fisiologia , Lipídeos/fisiologia , Proteínas de Membrana/metabolismo , Membranas/metabolismo , Modelos Biológicos , Modelos Teóricos , Biogênese de Organelas , Proteínas de Transferência de Fosfolipídeos/fisiologia
4.
Mol Reprod Dev ; 88(5): 371-375, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33821543

RESUMO

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.


Assuntos
Adenosina Trifosfatases/fisiologia , Fertilidade/fisiologia , Proteínas de Transferência de Fosfolipídeos/fisiologia , Espermatogênese/fisiologia , Proteínas rab de Ligação ao GTP/metabolismo , Adenosina Trifosfatases/química , Adenosina Trifosfatases/deficiência , Adenosina Trifosfatases/genética , Animais , Epididimo/ultraestrutura , Infertilidade Masculina/genética , Masculino , Lipídeos de Membrana/metabolismo , Camundongos , Camundongos Knockout , Fosfatidiletanolaminas/metabolismo , Fosfatidilserinas/metabolismo , Proteínas de Transferência de Fosfolipídeos/química , Proteínas de Transferência de Fosfolipídeos/deficiência , Proteínas de Transferência de Fosfolipídeos/genética , Domínios Proteicos , Testículo/ultraestrutura
5.
Cell Mol Immunol ; 18(3): 686-697, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33469162

RESUMO

One of the hallmarks of live cells is the asymmetric distribution of lipids across their plasma membrane. Changes in this asymmetry due to lipid "scrambling" result in phosphatidylserine exposure at the cell surface that is detected by annexin V staining. This alteration is observed during cell death processes such as apoptosis, and during physiological responses such as platelet degranulation and membrane repair. Previous studies have shown that activation of NK cells is accompanied by exposure of phosphatidylserine at the cell surface. While this response was thought to be indicative of ongoing NK cell death, it may also  reflect the regulation of NK cell activation in the absence of cell death. Herein, we found that NK cell activation was accompanied by rapid phosphatidylserine exposure to an extent proportional to the degree of NK cell activation. Through enforced expression of a lipid scramblase, we provided evidence that activation-induced lipid scrambling in NK cells is reversible and does not lead to cell death. In contrast, lipid scrambling attenuates NK cell activation. This response was accompanied by reduced cell surface expression of activating receptors such as 2B4, and by loss of binding of Src family protein tyrosine kinases Fyn and Lck to the inner leaflet of the plasma membrane. Hence, lipid scrambling during NK cell activation is, at least in part, a physiological response that reduces the NK cell activation level. This effect is due to the ability of lipid scrambling to alter the distribution of membrane-associated receptors and kinases required for NK cell activation.


Assuntos
Anoctaminas/fisiologia , Membrana Celular/metabolismo , Células Matadoras Naturais/imunologia , Proteína Tirosina Quinase p56(lck) Linfócito-Específica/metabolismo , Fosfatidilserinas/metabolismo , Proteínas de Transferência de Fosfolipídeos/fisiologia , Proteínas Proto-Oncogênicas c-fyn/metabolismo , Animais , Membrana Celular/imunologia , Células Matadoras Naturais/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout
6.
Cell Rep ; 33(13): 108570, 2020 12 29.
Artigo em Inglês | MEDLINE | ID: mdl-33378669

RESUMO

Calcium (Ca2+) is the primary stimulus for transmembrane protein 16 (TMEM16) Ca2+-activated chloride channels and phospholipid scramblases, which regulate important physiological processes ranging from smooth muscle contraction to blood coagulation and tumor progression. Binding of intracellular Ca2+ to two highly conserved orthosteric binding sites in transmembrane helices (TMs) 6-8 efficiently opens the permeation pathway formed by TMs 3-7. Recent structures of TMEM16K and TMEM16F scramblases revealed an additional Ca2+ binding site between TM2 and TM10, whose functional relevance remains unknown. Here, we report that Ca2+ binds with high affinity to the equivalent third Ca2+ site in TMEM16A to enhance channel activation. Our cadmium (Cd2+) metal bridging experiments reveal that the third Ca2+ site's conformational states can profoundly influence TMEM16A's opening. Our study thus confirms the existence of a third Ca2+ site in TMEM16A, defines its functional importance in channel gating, and provides insight into a long-range allosteric gating mechanism of TMEM16 channels and scramblases.


Assuntos
Anoctamina-1/fisiologia , Cálcio/metabolismo , Canais de Cloreto/fisiologia , Anoctamina-1/química , Sítios de Ligação , Cádmio/metabolismo , Membrana Celular/metabolismo , Eletrofisiologia/métodos , Células HEK293 , Humanos , Ativação do Canal Iônico , Transporte de Íons , Modelos Moleculares , Mutação , Proteínas de Transferência de Fosfolipídeos/fisiologia , Conformação Proteica , Domínios Proteicos
7.
Proc Natl Acad Sci U S A ; 117(43): 26907-26914, 2020 10 27.
Artigo em Inglês | MEDLINE | ID: mdl-33046656

RESUMO

The outer membrane (OM) of Gram-negative bacteria is a selective permeability barrier that allows uptake of nutrients while simultaneously protecting the cell from harmful compounds. The basic pathways and molecular machinery responsible for transporting lipopolysaccharides (LPS), lipoproteins, and ß-barrel proteins to the OM have been identified, but very little is known about phospholipid (PL) transport. To identify genes capable of affecting PL transport, we screened for genetic interactions with mlaA*, a mutant in which anterograde PL transport causes the inner membrane (IM) to shrink and eventually rupture; characterization of mlaA*-mediated lysis suggested that PL transport can occur via a high-flux diffusive flow mechanism. We found that YhdP, an IM protein involved in maintaining the OM permeability barrier, modulates the rate of PL transport during mlaA*-mediated lysis. Deletion of yhdP from mlaA* reduced the rate of IM transport to the OM by 50%, slowing shrinkage of the IM and delaying lysis. As a result, the weakened OM of ∆yhdP cells was further compromised and ruptured before the IM during mlaA*-mediated death. These findings demonstrate the existence of a high-flux diffusive pathway for PL flow in Escherichia coli that is modulated by YhdP.


Assuntos
Proteínas de Escherichia coli/fisiologia , Proteínas de Membrana/fisiologia , Proteínas de Transferência de Fosfolipídeos/fisiologia , Fosfolipídeos/metabolismo , Escherichia coli K12
8.
Adv Biol Regul ; 78: 100740, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-32992233

RESUMO

Cellular membranes are critical platforms for intracellular signaling that involve complex interfaces between lipids and proteins, and a web of interactions between a multitude of lipid metabolic pathways. Membrane lipids impart structural and functional information in this regulatory circuit that encompass biophysical parameters such as membrane thickness and fluidity, as well as chaperoning the interactions of protein binding partners. Phosphatidylinositol and its phosphorylated derivatives, the phosphoinositides, play key roles in intracellular membrane signaling, and these involvements are translated into an impressively diverse set of biological outcomes. The phosphatidylinositol transfer proteins (PITPs) are key regulators of phosphoinositide signaling. Found in a diverse array of organisms from plants, yeast and apicomplexan parasites to mammals, PITPs were initially proposed to be simple transporters of lipids between intracellular membranes. It now appears increasingly unlikely that the soluble versions of these proteins perform such functions within the cell. Rather, these serve to facilitate the activity of intrinsically biologically insufficient inositol lipid kinases and, in so doing, promote diversification of the biological outcomes of phosphoinositide signaling. The central engine for execution of such functions is the lipid exchange cycle that is a fundamental property of PITPs. How PITPs execute lipid exchange remains very poorly understood. Molecular dynamics simulation approaches are now providing the first atomistic insights into how PITPs, and potentially other lipid-exchange/transfer proteins, operate.


Assuntos
Inositol/metabolismo , Lipídeos de Membrana/metabolismo , Proteínas de Transferência de Fosfolipídeos/fisiologia , Fosfotransferases/metabolismo , Transdução de Sinais , Animais , Humanos
9.
J Plant Physiol ; 234-235: 94-97, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30690193

RESUMO

The plant patellin (PATL) proteins are yeast Sec14 protein (Sec14p)-like phosphatidylinositol transfer proteins (PITPs), which are widely distributed across the plant kingdom. The model plant Arabidopsis has six PATL members (designated as PATL1-PATL6). Accumulated evidence has indicated the involvement of Arabidopsis PATLs in various biological processes. This mini-review briefly summarizes our current knowledge on individual PATLs regarding their roles in plant development and stress tolerance regulation. The elucidation of PATLs' biological function in plants will provide new insights on plant membrane trafficking and its regulatory roles in either plant growth or environmental stress response signaling networks.


Assuntos
Proteínas de Transferência de Fosfolipídeos/fisiologia , Desenvolvimento Vegetal , Proteínas de Plantas/fisiologia , Plantas/genética , Estresse Fisiológico , Família Multigênica , Fosfolipídeos/metabolismo , Transdução de Sinais
10.
Sci Rep ; 8(1): 15394, 2018 10 18.
Artigo em Inglês | MEDLINE | ID: mdl-30337619

RESUMO

Excessive neutrophil degranulation is a common feature of many inflammatory disorders, including alpha-1 antitrypsin (AAT) deficiency. Our group has demonstrated that phospholipid transfer protein (PLTP) prevents neutrophil degranulation but serine proteases, which AAT inhibits, cleave PLTP in diseased airways. We propose to identify if airway PLTP activity can be restored by AAT augmentation therapy and how PLTP subdues degranulation of neutrophils in AAT deficient subjects. Airway PLTP activity was lower in AAT deficient patients but elevated in the airways of patients on augmentation therapy. Functional AAT protein (from PiMM homozygotes) prevented PLTP cleavage unlike its mutated ZZ variant (PiZZ). PLTP lowered leukotriene B4 induced degranulation of primary, secondary and tertiary granules from neutrophils from both groups (n = 14/group). Neutrophils isolated from Pltp knockout mice have enhance neutrophil degranulation. Both AAT and PLTP reduced neutrophil degranulation and superoxide production, possibly though their inhibition of the Src tyrosine kinase, Hck. Src kinase inhibitors saracatinib and dasatinib reduced neutrophil degranulation and superoxide production. Therefore, AAT protects PLTP from proteolytic cleavage and both AAT and PLTP mediate degranulation, possibly via Hck tyrosine kinase inhibition. Deficiency of AAT could contribute to reduced lung PLTP activity and elevated neutrophil signaling associated with lung disease.


Assuntos
Degranulação Celular/genética , Ativação de Neutrófilo/genética , Proteínas de Transferência de Fosfolipídeos/fisiologia , Proteínas Proto-Oncogênicas c-hck/metabolismo , alfa 1-Antitripsina/fisiologia , Idoso , Animais , Feminino , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Pessoa de Meia-Idade , Neutrófilos/fisiologia , Proteínas de Transferência de Fosfolipídeos/genética , Transdução de Sinais/genética , alfa 1-Antitripsina/genética , Deficiência de alfa 1-Antitripsina/genética , Deficiência de alfa 1-Antitripsina/metabolismo , Deficiência de alfa 1-Antitripsina/patologia
11.
Sci Rep ; 8(1): 10795, 2018 Jul 17.
Artigo em Inglês | MEDLINE | ID: mdl-30018401

RESUMO

P4-ATPases are a subfamily of P-type ATPases that flip phospholipids across membranes to generate lipid asymmetry, a property vital to many cellular processes. Mutations in several P4-ATPases have been linked to severe neurodegenerative and metabolic disorders. Most P4-ATPases associate with one of three accessory subunit isoforms known as CDC50A (TMEM30A), CDC50B (TMEM30B), and CDC50C (TMEM30C). To identify P4-ATPases that associate with CDC50A, in vivo, and determine their tissue distribution, we isolated P4-ATPases-CDC50A complexes from retina, brain, liver, testes, and kidney on a CDC50A immunoaffinity column and identified and quantified P4-ATPases from their tryptic peptides by mass spectrometry. Of the 12 P4-ATPase that associate with CDC50 subunits, 10 P4-ATPases were detected. Four P4-ATPases (ATP8A1, ATP11A, ATP11B, ATP11C) were present in all five tissues. ATP10D was found in low amounts in liver, brain, testes, and kidney, and ATP8A2 was present in significant amounts in retina, brain, and testes. ATP8B1 was detected only in liver, ATP8B3 and ATP10A only in testes, and ATP8B2 primarily in brain. We also show that ATP11A, ATP11B and ATP11C, like ATP8A1 and ATP8A2, selectively flip phosphatidylserine and phosphatidylethanolamine across membranes. These studies provide new insight into the tissue distribution, relative abundance, subunit interactions and substrate specificity of P4-ATPase-CDC50A complexes.


Assuntos
ATPases do Tipo-P/fisiologia , Proteínas de Transferência de Fosfolipídeos/fisiologia , Animais , Encéfalo/metabolismo , Rim/metabolismo , Fígado/metabolismo , Masculino , Espectrometria de Massas , Camundongos , ATPases do Tipo-P/metabolismo , Proteínas de Transferência de Fosfolipídeos/metabolismo , Proteômica , Retina/metabolismo , Testículo/metabolismo
12.
J Am Chem Soc ; 140(13): 4481-4484, 2018 04 04.
Artigo em Inglês | MEDLINE | ID: mdl-29558128

RESUMO

MurJ, the flippase that exports the bacterial cell wall monomer Lipid II to the periplasm, is a target for new antibiotics, which are desperately needed to treat Gram-negative infections. Quantitative methods to monitor MurJ activity are required to characterize inhibitors but are challenging to develop because the lipid-linked substrate is not chemically altered in a flippase reaction. Here we show that MurJ inhibition can be quantified by measuring the accumulation of intracellular Lipid II using a biotin-tagging strategy. We have exploited this assay to show that MurJ is inhibited in the presence of a compound that dissipates the membrane potential. By probing cysteine accessibility we have found that under this condition MurJ relaxes into an inactive, outward-facing conformation reminiscent of that targeted by the peptide antibiotic LysM. We conclude that membrane potential is required for MurJ function in E. coli, and we anticipate that the ability to accumulate this inactive conformation will lead to structures useful for inhibitor design.


Assuntos
Proteínas de Escherichia coli/fisiologia , Escherichia coli , Potenciais da Membrana , Proteínas de Transferência de Fosfolipídeos/fisiologia , Parede Celular/química , Parede Celular/fisiologia , Escherichia coli/metabolismo , Modelos Biológicos
13.
Dev Cell ; 44(6): 725-740.e4, 2018 03 26.
Artigo em Inglês | MEDLINE | ID: mdl-29587143

RESUMO

Phosphatidylinositol (PtdIns) transfer proteins (PITPs) stimulate PtdIns-4-P synthesis and signaling in eukaryotic cells, but to what biological outcomes such signaling circuits are coupled remains unclear. Herein, we show that two highly related StART-like PITPs, PITPNA and PITPNB, act in a redundant fashion to support development of the embryonic mammalian neocortex. PITPNA/PITPNB do so by driving PtdIns-4-P-dependent recruitment of GOLPH3, and likely ceramide transfer protein (CERT), to Golgi membranes with GOLPH3 recruitment serving to promote MYO18A- and F-actin-directed loading of the Golgi network to apical processes of neural stem cells (NSCs). We propose the primary role for PITP/PtdIns-4-P/GOLPH3/CERT signaling in NSC Golgi is not in regulating bulk membrane trafficking but in optimizing apically directed membrane trafficking and/or apical membrane signaling during neurogenesis.


Assuntos
Polaridade Celular , Complexo de Golgi/metabolismo , Células-Tronco Neurais/citologia , Neurogênese/fisiologia , Fosfatos de Fosfatidilinositol/metabolismo , Proteínas de Transferência de Fosfolipídeos/fisiologia , Animais , Membrana Celular/metabolismo , Células Cultivadas , Desenvolvimento Embrionário , Feminino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Miosinas/metabolismo , Células-Tronco Neurais/metabolismo , Fosfoproteínas/metabolismo , Transdução de Sinais
14.
PLoS Pathog ; 14(1): e1006848, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-29338048

RESUMO

Cell surface receptors for phosphatidylserine contribute to the entry of Ebola virus (EBOV) particles, indicating that the presence of phosphatidylserine in the envelope of EBOV is important for the internalization of EBOV particles. Phosphatidylserine is typically distributed in the inner layer of the plasma membrane in normal cells. Progeny virions bud from the plasma membrane of infected cells, suggesting that phosphatidylserine is likely flipped to the outer leaflet of the plasma membrane in infected cells for EBOV virions to acquire it. Currently, the intracellular dynamics of phosphatidylserine during EBOV infection are poorly understood. Here, we explored the role of XK-related protein (Xkr) 8, which is a scramblase responsible for exposure of phosphatidylserine in the plasma membrane of apoptotic cells, to understand its significance in phosphatidylserine-dependent entry of EBOV. We found that Xkr8 and transiently expressed EBOV glycoprotein GP often co-localized in intracellular vesicles and the plasma membrane. We also found that co-expression of GP and viral major matrix protein VP40 promoted incorporation of Xkr8 into ebolavirus-like particles (VLPs) and exposure of phosphatidylserine on their surface, although only a limited amount of phosphatidylserine was exposed on the surface of the cells expressing GP and/or VP40. Downregulating Xkr8 or blocking caspase-mediated Xkr8 activation did not affect VLP production, but they reduced the amount of phosphatidylserine on the VLPs and their uptake in recipient cells. Taken together, our findings indicate that Xkr8 is trafficked to budding sites via GP-containing vesicles, is incorporated into VLPs, and then promote the entry of the released EBOV to cells in a phosphatidylserine-dependent manner.


Assuntos
Ebolavirus/fisiologia , Interações Hospedeiro-Patógeno , Fosfatidilserinas/metabolismo , Proteínas de Transferência de Fosfolipídeos/fisiologia , Vírion/metabolismo , Animais , Chlorocebus aethiops , Células HEK293 , Doença pelo Vírus Ebola/metabolismo , Doença pelo Vírus Ebola/virologia , Humanos , Células Vero , Proteínas do Core Viral/metabolismo , Liberação de Vírus
15.
Proc Natl Acad Sci U S A ; 114(23): 6080-6085, 2017 06 06.
Artigo em Inglês | MEDLINE | ID: mdl-28533404

RESUMO

Duchenne muscular dystrophy (DMD) is a progressive muscle wasting disease caused by X-linked inherited mutations in the DYSTROPHIN (DMD) gene. Absence of dystrophin protein from the sarcolemma causes severe muscle degeneration, fibrosis, and inflammation, ultimately leading to cardiorespiratory failure and premature death. Although there are several promising strategies under investigation to restore dystrophin protein expression, there is currently no cure for DMD, and identification of genetic modifiers as potential targets represents an alternative therapeutic strategy. In a Brazilian golden retriever muscular dystrophy (GRMD) dog colony, two related dogs demonstrated strikingly mild dystrophic phenotypes compared with those typically observed in severely affected GRMD dogs despite lacking dystrophin. Microarray analysis of these "escaper" dogs revealed reduced expression of phosphatidylinositol transfer protein-α (PITPNA) in escaper versus severely affected GRMD dogs. Based on these findings, we decided to pursue investigation of modulation of PITPNA expression on dystrophic pathology in GRMD dogs, dystrophin-deficient sapje zebrafish, and human DMD myogenic cells. In GRMD dogs, decreased expression of Pitpna was associated with increased phosphorylated Akt (pAkt) expression and decreased PTEN levels. PITPNA knockdown by injection of morpholino oligonucleotides in sapje zebrafish also increased pAkt, rescued the abnormal muscle phenotype, and improved long-term sapje mutant survival. In DMD myotubes, PITPNA knockdown by lentiviral shRNA increased pAkt and increased myoblast fusion index. Overall, our findings suggest PIPTNA as a disease modifier that accords benefits to the abnormal signaling, morphology, and function of dystrophic skeletal muscle, and may be a target for DMD and related neuromuscular diseases.


Assuntos
Distrofia Muscular de Duchenne/metabolismo , Proteínas de Transferência de Fosfolipídeos/metabolismo , Proteínas de Transferência de Fosfolipídeos/fisiologia , Animais , Linhagem Celular , Modelos Animais de Doenças , Cães , Distrofina/genética , Distrofina/metabolismo , Humanos , Células Musculares/fisiologia , Fibras Musculares Esqueléticas/metabolismo , Músculo Esquelético/metabolismo , Distrofia Muscular Animal/genética , Distrofia Muscular de Duchenne/fisiopatologia , Mutação , Fosforilação , Proteínas Proto-Oncogênicas c-akt , Peixe-Zebra/metabolismo
16.
Cell Biol Int ; 41(4): 374-383, 2017 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-28150893

RESUMO

Lipid rafts are an initiation site for many different signals. Recently, we reported that an EGF domain in activated coagulation factor IX (EGF-F9) increases lipid raft formation and accelerates cell migration. However, the detailed mechanism is not well understood. This study aimed to evaluate the effects of EGF-F9 on the cell membrane. A431 cells (derived from human squamous cell carcinoma) were treated with recombinant EGF-F9. Cells were immunocytochemically stained with probes for lipid rafts or phosphatidylserine (PS). After 3 min of treatment with EGF-F9, cholera toxin subunit B (CTxB) binding domains emerged at the adhesive tips of filopodia. Subsequently, CTxB staining was observed on the filopodial shaft. Finally, large clusters of CTxB domains were observed at the edge of cell bodies. Markers for lipid rafts, such as caveolin-1 and a GPI anchored protein, co-localized with CTxB. Staining with annexin V and XII revealed that PS was exposed at the tips of filopodia, translocated on filopodial shafts, and co-localized with CTxB at the rafts. Immunocytochemistry showed that scramblase-1 protein was present at the filopodial tips. Our data indicates that EGF-F9 accelerates PS exposure around the filopodial adhesion complex and induces clustering of lipid rafts in the cell body. PS exposure is thought to occur on cells undergoing apoptosis. Further study of the function of the EGF-F9 motif in mediating signal transduction is necessary because it is shared by a number of proteins.


Assuntos
Fator IX/fisiologia , Fosfatidilserinas/metabolismo , Apoptose , Linhagem Celular Tumoral , Fator de Crescimento Epidérmico/fisiologia , Humanos , Proteínas de Transferência de Fosfolipídeos/fisiologia , Domínios Proteicos
17.
Mol Biol Cell ; 28(3): 452-462, 2017 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-27932490

RESUMO

ATP8A2 is a P4-ATPase that flips phosphatidylserine and phosphatidylethanolamine across cell membranes. This generates membrane phospholipid asymmetry, a property important in many cellular processes, including vesicle trafficking. ATP8A2 deficiency causes severe neurodegenerative diseases. We investigated the role of the C-terminus of ATP8A2 in its expression, subcellular localization, interaction with its subunit CDC50A, and function as a phosphatidylserine flippase. C-terminal deletion mutants exhibited a reduced tendency to solubilize in mild detergent and exit the endoplasmic reticulum. The solubilized protein, however, assembled with CDC50A and displayed phosphatidylserine flippase activity. Deletion of the C-terminal 33 residues resulted in reduced phosphatidylserine-dependent ATPase activity, phosphatidylserine flippase activity, and neurite extension in PC12 cells. These reduced activities were reversed with 60- and 80-residue C-terminal deletions. Unlike the yeast P4-ATPase Drs2, ATP8A2 is not regulated by phosphoinositides but undergoes phosphorylation on the serine residue within a CaMKII target motif. We propose a model in which the C-terminus of ATP8A2 consists of an autoinhibitor domain upstream of the C-terminal 33 residues and an anti-autoinhibitor domain at the extreme C-terminus. The latter blocks the inhibitory activity of the autoinhibitor domain. We conclude that the C-terminus plays an important role in the efficient folding and regulation of ATP8A2.


Assuntos
Adenosina Trifosfatases/genética , Adenosina Trifosfatases/metabolismo , Adenosina Trifosfatases/fisiologia , Proteínas de Transferência de Fosfolipídeos/genética , Proteínas de Transferência de Fosfolipídeos/metabolismo , Proteínas de Transferência de Fosfolipídeos/fisiologia , Animais , Transporte Biológico , Membrana Celular/metabolismo , Células HEK293 , Humanos , Proteínas de Membrana/metabolismo , Proteínas de Membrana/fisiologia , Células PC12 , Fosfatidiletanolaminas/metabolismo , Fosfatidilserinas/metabolismo , Fosfolipídeos/metabolismo , Domínios Proteicos , Dobramento de Proteína , Transporte Proteico , Ratos
18.
Proc Natl Acad Sci U S A ; 113(49): 14025-14030, 2016 12 06.
Artigo em Inglês | MEDLINE | ID: mdl-27872310

RESUMO

Our understanding of membranes and membrane lipid function has lagged far behind that of nucleic acids and proteins, largely because it is difficult to manipulate cellular membrane lipid composition. To help solve this problem, we show that methyl-α-cyclodextrin (MαCD)-catalyzed lipid exchange can be used to maximally replace the sphingolipids and phospholipids in the outer leaflet of the plasma membrane of living mammalian cells with exogenous lipids, including unnatural lipids. In addition, lipid exchange experiments revealed that 70-80% of cell sphingomyelin resided in the plasma membrane outer leaflet; the asymmetry of metabolically active cells was similar to that previously defined for erythrocytes, as judged by outer leaflet lipid composition; and plasma membrane outer leaflet phosphatidylcholine had a significantly lower level of unsaturation than phosphatidylcholine in the remainder of the cell. The data also provided a rough estimate for the total cellular lipids residing in the plasma membrane (about half). In addition to such lipidomics applications, the exchange method should have wide potential for investigations of lipid function and modification of cellular behavior by modification of lipids.


Assuntos
Metabolismo dos Lipídeos/fisiologia , Proteínas de Transferência de Fosfolipídeos/metabolismo , alfa-Ciclodextrinas/metabolismo , Células A549/metabolismo , Animais , Membrana Celular/metabolismo , Membrana Celular/fisiologia , Ciclodextrinas/metabolismo , Ciclodextrinas/farmacologia , Humanos , Bicamadas Lipídicas/metabolismo , Lipídeos/fisiologia , Espectrometria de Massas , Lipídeos de Membrana/metabolismo , Fosfatidilcolinas/metabolismo , Proteínas de Transferência de Fosfolipídeos/fisiologia , Fosfolipídeos/metabolismo , Esfingolipídeos/metabolismo , Esfingomielinas , alfa-Ciclodextrinas/farmacologia
19.
Mol Biol Cell ; 27(24): 3883-3893, 2016 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-27733620

RESUMO

Type IV P-type ATPases (P4-ATPases) are phospholipid flippases that translocate phospholipids from the exoplasmic (or luminal) to the cytoplasmic leaflet of lipid bilayers. In Saccharomyces cerevisiae, P4-ATPases are localized to specific subcellular compartments and play roles in compartment-mediated membrane trafficking; however, roles of mammalian P4-ATPases in membrane trafficking are poorly understood. We previously reported that ATP9A, one of 14 human P4-ATPases, is localized to endosomal compartments and the Golgi complex. In this study, we found that ATP9A is localized to phosphatidylserine (PS)-positive early and recycling endosomes, but not late endosomes, in HeLa cells. Depletion of ATP9A delayed the recycling of transferrin from endosomes to the plasma membrane, although it did not affect the morphology of endosomal structures. Moreover, depletion of ATP9A caused accumulation of glucose transporter 1 in endosomes, probably by inhibiting their recycling. By contrast, depletion of ATP9A affected neither the early/late endosomal transport and degradation of epidermal growth factor (EGF) nor the transport of Shiga toxin B fragment from early/recycling endosomes to the Golgi complex. Therefore ATP9A plays a crucial role in recycling from endosomes to the plasma membrane.


Assuntos
Endossomos/metabolismo , Proteínas de Transferência de Fosfolipídeos/metabolismo , Proteínas de Transferência de Fosfolipídeos/fisiologia , Adenosina Trifosfatases/metabolismo , Transporte Biológico , Membrana Celular/metabolismo , Endossomos/fisiologia , Complexo de Golgi/metabolismo , Células HeLa , Humanos , Bicamadas Lipídicas/metabolismo , Proteínas de Membrana/metabolismo , Fosfatidilserinas/metabolismo , Fosfolipídeos/metabolismo , Transporte Proteico , Vesículas Transportadoras/metabolismo
20.
Inflammation ; 39(6): 1972-1980, 2016 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-27596005

RESUMO

To investigate the effect of phospholipid transfer protein (PLTP) on the cigarette smoke extract (CSE)-induced production of interleukin-8 (IL-8) in human pulmonary epithelial cells, male Wistar rats were exposed to air and cigarette smoke (n = 10/exposure) for 6 h/day on three consecutive days. Their lungs were sectioned and bronchoalveolar lavage fluid (BALF) examined. The expression of PLTP and IL-8 in the lung was detected immunohistochemically. Lung injury was accompanied by the upregulation of PLTP and IL-8 in the CSE-exposed rat model, and the number of white blood cells in the BALF was significantly increased compared with those of the controls. Both neutrophils and macrophages were clearly increased. Human alveolar epithelial cells (A549) and human bronchial epithelial cells (HBECs) were treated with different concentrations of CSE for various times. The cells were also transfected with small interfering RNA directed against PLTP, and U0126, an inhibitor of the ERK1/2 pathway, was administered before CSE exposure. The expression of PLTP and IL-8 mRNAs and PLTP, IL-8, total ERK, and phosphorylated ERK proteins was analyzed. The expression of IL-8 and phosphorylated ERK was significantly increased in A549 cells and HBECs after CSE stimulation, and CSE upregulated the expression of PLTP in A549 cells. In contrast, CSE inhibited the expression of PLTP in HBECs. The CSE-induced expression of IL-8 and p-ERK was significantly increased by the knockdown of PLTP. Therefore, PLTP may regulate CSE-induced IL-8 expression via the ERK1/2 signaling pathway in human pulmonary epithelial cells.


Assuntos
Células Epiteliais/metabolismo , Interleucina-8/biossíntese , Proteínas de Transferência de Fosfolipídeos/fisiologia , Fumaça , Células A549 , Animais , Brônquios/patologia , Humanos , Sistema de Sinalização das MAP Quinases , Masculino , Fosforilação , Ratos , Ratos Wistar
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...