Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Int J Mol Sci ; 24(4)2023 Feb 14.
Artigo em Inglês | MEDLINE | ID: mdl-36835206

RESUMO

Kinesin family motors are microtubule (MT)-stimulated ATPases known best as transporters of cellular cargoes through the cytoplasm, regulators of MT dynamics, organizers of the mitotic spindle, and for insuring equal division of DNA during mitosis. Several kinesins have also been shown to regulate transcription by interacting with transcriptional cofactors and regulators, nuclear receptors, or with specific promotor elements on DNA. We previously showed that an LxxLL nuclear receptor box motif in the kinesin-2 family motor KIF17 mediates binding to the orphan nuclear receptor estrogen related receptor alpha (ERR1) and is responsible for the suppression of ERR1-dependent transcription by KIF17. Analysis of all kinesin family proteins revealed that multiple kinesins contain this LxxLL motif, raising the question as to whether additional kinesin motors contribute to the regulation of ERR1. In this study, we interrogate the effects of multiple kinesins with LxxLL motifs on ERR1-mediated transcription. We demonstrate that the kinesin-3 family motor KIF1B contains two LxxLL motifs, one of which binds to ERR1. In addition, we show that expression of a KIF1B fragment containing this LxxLL motif inhibits ERR1-dependent transcription by regulating nuclear entry of ERR1. We also provide evidence that the effects of expressing the KIF1B-LxxLL fragment on ERR1 activity are mediated by a mechanism distinct from that of KIF17. Since LxxLL domains are found in many kinesins, our data suggest an expanded role for kinesins in nuclear receptor mediated transcriptional regulation.


Assuntos
Regulação da Expressão Gênica , Cinesinas , Mitose , Receptores de Estrogênio , Núcleo Celular/metabolismo , Cinesinas/metabolismo , Microtúbulos/metabolismo , Receptores de Estrogênio/genética , Receptores de Estrogênio/metabolismo , Motivos de Aminoácidos/genética
2.
J Exp Med ; 216(11): 2582-2601, 2019 11 04.
Artigo em Inglês | MEDLINE | ID: mdl-31395618

RESUMO

Transendothelial migration (TEM) of leukocytes across the endothelium is critical for inflammation. In the endothelium, TEM requires the coordination of membrane movements and cytoskeletal interactions, including, prominently, recruitment of the lateral border recycling compartment (LBRC). The scaffold protein IQGAP1 was recently identified in a screen for LBRC-interacting proteins. Knockdown of endothelial IQGAP1 disrupted the directed movement of the LBRC and substantially reduced leukocyte TEM. Expression of truncated IQGAP1 constructs demonstrated that the calponin homology domain is required for IQGAP1 localization to endothelial borders and that the IQ domain, on the same IQGAP1 polypeptide, is required for its function in TEM. This is the first reported function of IQGAP1 requiring two domains to be present on the same polypeptide. Additionally, we show for the first time that IQGAP1 in the endothelium is required for efficient TEM in vivo. These findings reveal a novel function for IQGAP1 and demonstrate that IQGAP1 in endothelial cells facilitates TEM by directing the LBRC to the site of TEM.


Assuntos
Células Endoteliais/metabolismo , Leucócitos/metabolismo , Migração Transendotelial e Transepitelial , Proteínas Ativadoras de ras GTPase/metabolismo , Citoesqueleto de Actina/metabolismo , Animais , Antígenos CD , Caderinas , Células Cultivadas , Humanos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Microscopia de Fluorescência , Microtúbulos/metabolismo , Transporte Proteico , Interferência de RNA , Proteínas Ativadoras de ras GTPase/genética
3.
J Cell Sci ; 131(11)2018 06 11.
Artigo em Inglês | MEDLINE | ID: mdl-29748377

RESUMO

Epithelial cells require a precise intracellular transport and sorting machinery to establish and maintain their polarized architecture. This machinery includes ß-galactoside-binding galectins for targeting of glycoprotein to the apical membrane. Galectin-3 sorts cargo destined for the apical plasma membrane into vesicular carriers. After delivery of cargo to the apical milieu, galectin-3 recycles back into sorting organelles. We analysed the role of galectin-3 in the polarized distribution of ß1-integrin in MDCK cells. Integrins are located primarily at the basolateral domain of epithelial cells. We demonstrate that a minor pool of ß1-integrin interacts with galectin-3 at the apical plasma membrane. Knockdown of galectin-3 decreases apical delivery of ß1-integrin. This loss is restored by supplementation with recombinant galectin-3 and galectin-3 overexpression. Our data suggest that galectin-3 targets newly synthesized ß1-integrin to the apical membrane and promotes apical delivery of ß1-integrin internalized from the basolateral membrane. In parallel, knockout of galectin-3 results in a reduction in cell proliferation and an impairment in proper cyst development. Our results suggest that galectin-3 modulates the surface distribution of ß1-integrin and affects the morphogenesis of polarized cells.


Assuntos
Polaridade Celular , Células Epiteliais/metabolismo , Galectina 3/metabolismo , Integrina beta1/metabolismo , Animais , Membrana Celular/genética , Membrana Celular/metabolismo , Proliferação de Células , Cães , Células Epiteliais/citologia , Galectina 3/genética , Integrina beta1/genética , Células Madin Darby de Rim Canino , Transporte Proteico
5.
Artigo em Inglês | MEDLINE | ID: mdl-28264820

RESUMO

Epithelial cells play a key role in insuring physiological homeostasis by acting as a barrier between the outside environment and internal organs. They are also responsible for the vectorial transport of ions and fluid essential to the function of many organs. To accomplish these tasks, epithelial cells must generate an asymmetrically organized plasma membrane comprised of structurally and functionally distinct apical and basolateral membranes. Adherent and occluding junctions, respectively, anchor cells within a layer and prevent lateral diffusion of proteins in the outer leaflet of the plasma membrane and restrict passage of proteins and solutes through intercellular spaces. At a fundamental level, the establishment and maintenance of epithelial polarity requires that signals initiated at cell-substratum and cell-cell adhesions are transmitted appropriately and dynamically to the cytoskeleton, to the membrane-trafficking machinery, and to the regulation of occluding and adherent junctions. Rigorous descriptive and mechanistic studies published over the last 50 years have provided great detail to our understanding of epithelial polarization. Yet still, critical early steps in morphogenesis are not yet fully appreciated. In this review, we discuss how cytoskeletal motor proteins, primarily kinesins, contribute to coordinated modification of microtubule and actin arrays, formation and remodeling of cell adhesions to targeted membrane trafficking, and to initiating the formation and expansion of an apical lumen.


Assuntos
Polaridade Celular , Células Epiteliais/fisiologia , Microtúbulos/fisiologia , Animais , Adesão Celular , Citoesqueleto/fisiologia , Humanos , Morfogênese
6.
J Mol Biol ; 429(24): 3836-3849, 2017 12 08.
Artigo em Inglês | MEDLINE | ID: mdl-29079481

RESUMO

Heterotrimeric G-proteins are essential cellular signal transducers. One of the G-proteins, Gα13, is critical for actin cytoskeletal reorganization, cell migration, cell proliferation, and apoptosis. Previously, we have shown that Gα13 is essential for both G-protein-coupled receptor and receptor tyrosine kinase-induced actin cytoskeletal reorganization such as dynamic dorsal ruffle turnover and cell migration. However, the mechanism by which Gα13 signals to actin cytoskeletal reorganization is not completely understood. Here we show that Gα13 directly interacts with Abl tyrosine kinase, which is a critical regulator of actin cytoskeleton. This interaction is critical for Gα13-induced dorsal ruffle turnover, endothelial cell remodeling, and cell migration. Our data uncover a new molecular signaling pathway by which Gα13 controls actin cytoskeletal reorganization.


Assuntos
Citoesqueleto de Actina/metabolismo , Movimento Celular/fisiologia , Embrião de Mamíferos/metabolismo , Fibroblastos/metabolismo , Subunidades alfa G12-G13 de Proteínas de Ligação ao GTP/metabolismo , Proteínas Oncogênicas v-abl/metabolismo , Animais , Células Cultivadas , Embrião de Mamíferos/citologia , Fibroblastos/citologia , Subunidades alfa G12-G13 de Proteínas de Ligação ao GTP/genética , Camundongos , Camundongos Knockout , Proteínas Oncogênicas v-abl/genética , Transdução de Sinais , Esferoides Celulares , Cicatrização
7.
Oncotarget ; 8(31): 50359-50375, 2017 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-28881568

RESUMO

Estrogen-related receptor alpha (ERR1) is an orphan nuclear receptor that can bind transcriptional co-activators constitutively. ERR1 expression correlates with poor patient outcomes in breast cancer, heightening interest in this nuclear receptor as a therapeutic target. Because ERR1 has no known regulatory ligand, a major challenge in targeting its activity is to find cellular or synthetic modulators of its function. We identified an interaction between ERR1 and KIF17, a kinesin-2 family microtubule motor, in a yeast-2-hybrid screen. We confirmed the interaction using in vitro biochemical assays and determined that binding is mediated by the ERR1 ligand-binding/AF2 domain and the KIF17 C-terminal tail. Expression of KIF17 tail domain in either ER-negative or ER-positive breast cancer epithelial cells attenuated nuclear accumulation of newly synthesized ERR1 and inhibited ERR1 transcriptional activity. Conversely, ERR1 transcriptional activity was elevated significantly in KIF17 knock-out cells. Sequence analysis of the KIF17 tail domain revealed it contains a nuclear receptor box with a conserved LXXLL motif found in transcriptional co-activators. Expression of a 12 amino-acid peptide containing this motif was sufficient to inhibit ERR1 transcriptional activity and cell invasion, while deletion of this region from the KIF17 tail resulted in increased ERR1 activity. Together, these data suggest KIF17 modifies ERR1 function by two possible, non-exclusive mechanisms: (i) by regulating nuclear-cytoplasmic distribution or (ii) by competing with transcriptional co-activators for binding to ERR1. Thus targeting the ERR1-KIF17 interaction has potential as a novel strategy for treating breast cancer.

8.
J Cell Sci ; 129(5): 957-70, 2016 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-26759174

RESUMO

The kinesin KIF17 localizes at microtubule plus-ends where it contributes to regulation of microtubule stabilization and epithelial polarization. We now show that KIF17 localizes at cell-cell adhesions and that KIF17 depletion inhibits accumulation of actin at the apical pole of cells grown in 3D organotypic cultures and alters the distribution of actin and E-cadherin in cells cultured in 2D on solid supports. Overexpression of full-length KIF17 constructs or truncation mutants containing the N-terminal motor domain resulted in accumulation of newly incorporated GFP-actin into junctional actin foci, cleared E-cadherin from cytoplasmic vesicles and stabilized cell-cell adhesions to challenge with calcium depletion. Expression of these KIF17 constructs also increased cellular levels of active RhoA, whereas active RhoA was diminished in KIF17-depleted cells. Inhibition of RhoA or its effector ROCK, or expression of LIMK1 kinase-dead or activated cofilin(S3A) inhibited KIF17-induced junctional actin accumulation. Interestingly, KIF17 activity toward actin depends on the motor domain but is independent of microtubule binding. Together, these data show that KIF17 can modify RhoA-GTPase signaling to influence junctional actin and the stability of the apical junctional complex of epithelial cells.


Assuntos
Citoesqueleto de Actina/metabolismo , Células Epiteliais/fisiologia , Cinesinas/fisiologia , Proteína rhoA de Ligação ao GTP/metabolismo , Fatores de Despolimerização de Actina/metabolismo , Animais , Antígenos CD , Caderinas/metabolismo , Adesão Celular , Cães , Células Epiteliais/ultraestrutura , Quinases Lim/metabolismo , Células Madin Darby de Rim Canino , Microtúbulos/metabolismo , Ligação Proteica , Transporte Proteico , Transdução de Sinais , Quinases Associadas a rho/metabolismo
9.
J Cell Biol ; 206(6): 779-97, 2014 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-25225338

RESUMO

Mechanisms by which microtubule plus ends interact with regions of cell-cell contact during tissue development and morphogenesis are not fully understood. We characterize a previously unreported interaction between the microtubule binding protein end-binding 1 (EB1) and the desmosomal protein desmoplakin (DP), and demonstrate that DP-EB1 interactions enable DP to modify microtubule organization and dynamics near sites of cell-cell contact. EB1 interacts with a region of the DP N terminus containing a hotspot for pathogenic mutations associated with arrhythmogenic cardiomyopathy (AC). We show that a subset of AC mutations, in addition to a mutation associated with skin fragility/woolly hair syndrome, impair gap junction localization and function by misregulating DP-EB1 interactions and altering microtubule dynamics. This work identifies a novel function for a desmosomal protein in regulating microtubules that affect membrane targeting of gap junction components, and elucidates a mechanism by which DP mutations may contribute to the development of cardiac and cutaneous diseases.


Assuntos
Displasia Arritmogênica Ventricular Direita/genética , Conexina 43/metabolismo , Desmoplaquinas/metabolismo , Proteínas Associadas aos Microtúbulos/metabolismo , Microtúbulos/metabolismo , Animais , Células COS , Comunicação Celular , Linhagem Celular , Chlorocebus aethiops , Demecolcina/farmacologia , Desmogleína 2/genética , Desmoplaquinas/genética , Desmossomos/fisiologia , Junções Comunicantes/genética , Junções Comunicantes/patologia , Células HEK293 , Humanos , Morfogênese , Mutação , Interferência de RNA , RNA Interferente Pequeno , Ratos , Ratos Sprague-Dawley , Moduladores de Tubulina/farmacologia
10.
J Cell Biol ; 195(7): 1185-203, 2011 Dec 26.
Artigo em Inglês | MEDLINE | ID: mdl-22184201

RESUMO

The desmosomal cadherins, desmogleins (Dsgs) and desmocollins (Dscs), comprise the adhesive core of intercellular junctions known as desmosomes. Although these adhesion molecules are known to be critical for tissue integrity, mechanisms that coordinate their trafficking into intercellular junctions to regulate their proper ratio and distribution are unknown. We demonstrate that Dsg2 and Dsc2 both exhibit microtubule-dependent transport in epithelial cells but use distinct motors to traffic to the plasma membrane. Functional interference with kinesin-1 blocked Dsg2 transport, resulting in the assembly of Dsg2-deficient junctions with minimal impact on distribution of Dsc2 or desmosomal plaque components. In contrast, inhibiting kinesin-2 prevented Dsc2 movement and decreased its plasma membrane accumulation without affecting Dsg2 trafficking. Either kinesin-1 or -2 deficiency weakened intercellular adhesion, despite the maintenance of adherens junctions and other desmosome components at the plasma membrane. Differential regulation of desmosomal cadherin transport could provide a mechanism to tailor adhesion strength during tissue morphogenesis and remodeling.


Assuntos
Desmocolinas/metabolismo , Desmogleína 2/metabolismo , Desmossomos/metabolismo , Cinesinas/metabolismo , Membrana Celular/metabolismo , Humanos , Junções Intercelulares/metabolismo , Cinesinas/deficiência , Microtúbulos/metabolismo , Ligação Proteica , Transporte Proteico , Células Tumorais Cultivadas , Gravação em Vídeo
11.
J Cell Biol ; 190(3): 443-60, 2010 Aug 09.
Artigo em Inglês | MEDLINE | ID: mdl-20696710

RESUMO

Epithelial polarization is associated with selective stabilization and reorganization of microtubule (MT) arrays. However, upstream events and downstream consequences of MT stabilization during epithelial morphogenesis are still unclear. We show that the anterograde kinesin KIF17 localizes to MT plus ends, stabilizes MTs, and affects epithelial architecture. Targeting of KIF17 to plus ends of growing MTs requires kinesin motor activity and interaction with EB1. In turn, KIF17 participates in localizing adenomatous polyposis coli (APC) to the plus ends of a subset of MTs. We found that KIF17 affects MT dynamics, polymerization rates, and MT plus end stabilization to generate posttranslationally acetylated MTs. Depletion of KIF17 from cells growing in three-dimensional matrices results in aberrant epithelial cysts that fail to generate a single central lumen and to polarize apical markers. These findings implicate KIF17 in MT stabilization events that contribute to epithelial polarization and morphogenesis.


Assuntos
Proteína da Polipose Adenomatosa do Colo/metabolismo , Células Epiteliais/metabolismo , Cinesinas/metabolismo , Proteínas Associadas aos Microtúbulos/metabolismo , Microtúbulos/metabolismo , Morfogênese , Animais , Células CACO-2 , Células Cultivadas , Humanos , Camundongos
12.
J Cell Sci ; 123(Pt 10): 1732-41, 2010 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-20427314

RESUMO

A key process during epithelial polarization involves establishment of polarized transport routes from the Golgi to distinct apical and basolateral membrane domains. To do this, the machinery involved in selective trafficking must be regulated during differentiation. Our previous studies showed that KIF5B selectively transports vesicles containing p75-neurotrophin receptors to the apical membrane of polarized, but not non-polarized MDCK cells. To identify the kinesin(s) responsible for p75 trafficking in non-polarized MDCK cells we expressed KIF-specific dominant-negative constructs and assayed for changes in post-Golgi transport of p75 by time-lapse fluorescence microscopy. Overexpression of the tail domains of kinesin-3 family members that contain a C-terminal pleckstrin homology (PH) domain, KIF1A or KIF1Bbeta, attenuated the rate of p75 exit from the Golgi in non-polarized MDCK cells but not in polarized cells. Analysis of p75 post-Golgi transport in cells expressing KIF1A or KIF1Bbeta with their PH domains deleted revealed that vesicle transport by these motors depends on the PH domains. Furthermore, purified KIF1A and KIF1Bbeta tails interact with p75 vesicles and these interactions require the PH domain. Knockdown of canine KIF1A also inhibited exit of p75 from the Golgi, and this was rescued by expression of human KIF1A. Together these data demonstrate that post-Golgi transport of p75 in non-polarized epithelial cells is mediated by kinesin-3 family motors in a PH-domain-dependent process.


Assuntos
Células Epiteliais/metabolismo , Cinesinas/metabolismo , Receptor de Fator de Crescimento Neural/metabolismo , Animais , Linhagem Celular , Polaridade Celular , Clonagem Molecular , Cães , Células Epiteliais/patologia , Complexo de Golgi/metabolismo , Concentração de Íons de Hidrogênio , Cinesinas/genética , Cinesinas/isolamento & purificação , Microdomínios da Membrana/metabolismo , Estrutura Terciária de Proteína/genética , Transporte Proteico/genética , RNA Interferente Pequeno/genética , Transgenes/genética
13.
J Virol ; 82(20): 9937-50, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18684836

RESUMO

Retroviral Gag proteins are synthesized as soluble, myristoylated precursors that traffic to the plasma membrane and promote viral particle production. The intracellular transport of human immunodeficiency virus type 1 (HIV-1) Gag to the plasma membrane remains poorly understood, and cellular motor proteins responsible for Gag movement are not known. Here we show that disrupting the function of KIF4, a kinesin family member, slowed temporal progression of Gag through its trafficking intermediates and inhibited virus-like particle production. Knockdown of KIF4 also led to increased Gag degradation, resulting in reduced intracellular Gag protein levels; this phenotype was rescued by reintroduction of KIF4. When KIF4 function was blocked, Gag transiently accumulated in discrete, perinuclear, nonendocytic clusters that colocalized with endogenous KIF4, with Ubc9, an E2 SUMO-1 conjugating enzyme, and with SUMO. These studies identify a novel transit station through which Gag traffics en route to particle assembly and highlight the importance of KIF4 in regulating HIV-1 Gag trafficking and stability.


Assuntos
HIV-1/metabolismo , Cinesinas/metabolismo , Produtos do Gene gag do Vírus da Imunodeficiência Humana/metabolismo , Animais , Células COS , Chlorocebus aethiops , Citoplasma/metabolismo , HIV-1/genética , Humanos , Cinesinas/genética , Transporte Proteico/fisiologia , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Proteína SUMO-1/genética , Proteína SUMO-1/metabolismo , Enzimas de Conjugação de Ubiquitina/genética , Enzimas de Conjugação de Ubiquitina/metabolismo , Produtos do Gene gag do Vírus da Imunodeficiência Humana/genética
14.
Dev Cell ; 13(4): 511-22, 2007 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17925227

RESUMO

Microtubule-based vesicular transport is well documented in epithelial cells, but the specific motors involved and their regulation during polarization are largely unknown. We demonstrate that KIF5B mediates post-Golgi transport of an apical protein in epithelial cells, but only after polarity has developed. Time-lapse imaging of EB1-GFP in polarized MDCK cells showed microtubule plus ends growing toward the apical membrane, implying that plus end-directed N-kinesins might be used to transport apical proteins. Indeed, time-lapse microscopy revealed that expression of a KIF5B dominant negative or microinjection of function-blocking KIF5 antibodies inhibited selectively post-Golgi transport of the apical marker, p75-GFP, after polarization of MDCK cells. Expression of other KIF dominant negatives did not alter p75-GFP trafficking. Immunoprecipitation experiments demonstrated an interaction between KIF5B and p75-GFP in polarized, but not in subconfluent, MDCK cells. Our results demonstrate that apical protein transport depends on selective microtubule motors and that epithelial cells switch kinesins for post-Golgi transport during acquisition of polarity.


Assuntos
Membrana Celular/metabolismo , Polaridade Celular/fisiologia , Cinesinas/fisiologia , Proteínas de Membrana/metabolismo , Microtúbulos/fisiologia , Animais , Linhagem Celular , Cães , Células Epiteliais/metabolismo , Proteínas de Fluorescência Verde/genética , Humanos , Proteínas de Membrana/genética , Transporte Proteico , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo
15.
J Biol Chem ; 281(43): 32660-7, 2006 Oct 27.
Artigo em Inglês | MEDLINE | ID: mdl-16943201

RESUMO

Growth factors induce massive actin cytoskeletal remodeling in cells. These reorganization events underlie various cellular responses such as cell migration and morphological changes. One major form of actin reorganization is the formation and disassembly of dorsal ruffles (also named waves, dorsal rings, or circular ruffles). Dorsal ruffles are involved in physiological functions including cell migration, invasion, macropinocytosis, plasma membrane recycling, and others. Growth factors initiate rapid formation (within 5 min) of circular membrane ruffles, and these ruffles move along the dorsal side of the cells, constrict, close, and eventually disassemble ( approximately 20 min). Considerable attention has been devoted to the mechanism by which growth factors induce the formation of dorsal ruffles. However, little is known of the mechanism by which these ruffles are disassembled. Here we have shown that G proteins G(12) and G(13) control the rate of disassembly of dorsal ruffles. In Galpha(12)(-/-)Galpha(13)(-/-) fibroblast cells, dorsal ruffles induced by growth factor treatment remain visible substantially longer ( approximately 60 min) than in wild-type cells, whereas the rate of formation of these ruffles was the same with or without Galpha(12) and Galpha(13). Thus, Galpha(12)/Galpha(13) critically regulate dorsal ruffle turnover.


Assuntos
Membrana Celular/efeitos dos fármacos , Membrana Celular/fisiologia , Fibroblastos/efeitos dos fármacos , Subunidades alfa G12-G13 de Proteínas de Ligação ao GTP/fisiologia , Fator de Crescimento Derivado de Plaquetas/farmacologia , Actinas/metabolismo , Animais , Becaplermina , Células Cultivadas , Citoesqueleto/metabolismo , Feto/citologia , Fibroblastos/citologia , Fibroblastos/metabolismo , Recuperação de Fluorescência Após Fotodegradação , Subunidades alfa G12-G13 de Proteínas de Ligação ao GTP/deficiência , Proteínas de Fluorescência Verde/metabolismo , Imuno-Histoquímica , Camundongos , Microscopia de Fluorescência , Microscopia de Vídeo , Mutação , Células NIH 3T3 , Proteínas Proto-Oncogênicas c-sis , Fatores de Tempo
16.
J Biol Chem ; 279(42): 43411-8, 2004 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-15302870

RESUMO

Annexin 2 is a profibrinolytic co-receptor for plasminogen and tissue plasminogen activator that stimulates activation of the major fibrinolysin, plasmin, at cell surfaces. In human subjects, overexpression of annexin 2 in acute promyelocytic leukemia leads to a bleeding diathesis reflective of excessive cell surface annexin 2-dependent generation of plasmin (Menell, J. S., Cesarman, G. M., Jacovina, A. T., McLaughlin, M. A., Lev, E. A., and Hajjar, K. A. (1999) N. Engl. J. Med. 340, 994-1004). In addition, mice completely deficient in annexin 2 display fibrin accumulation within blood vessels and impaired clearance of injury-induced thrombi (Ling Q., Jacovina, A.T., Deora, A.B., Febbraio, M., Simantov, R., Silverstein, R. L., Hempstead, B. L., Mark, W., and Hajjar, K. A. (2004) J. Clin. Investig. 113, 38-48). Here, we show that endothelial cell annexin 2, a protein that lacks a typical signal peptide, translocates from the cytoplasm to the extracytoplasmic plasma membrane in response to brief temperature stress both in vitro and in vivo in the absence of cell death or cell lysis. This regulated response is independent of new protein or mRNA synthesis and does not require the classical endoplasmic reticulum-Golgi pathway. Temperature stress-induced annexin 2 translocation is dependent on both expression of protein p11 (S100A10) and tyrosine phosphorylation of annexin 2 because annexin 2 release is completely eliminated on depletion of p11, inactivation of tyrosine kinase, or mutation of tyrosine 23. Translocation of annexin 2 to the cell surface dramatically increases tissue plasminogen activator-dependent plasminogen activation potential and may represent a novel stress-induced protein secretion pathway.


Assuntos
Anexina A2/metabolismo , Membrana Celular/metabolismo , Endotélio Vascular/metabolismo , Proteínas S100/metabolismo , Anexina A2/genética , Apoptose , Células Cultivadas , Clonagem Molecular , DNA Complementar , Humanos , Cinética , Fosforilação , Transporte Proteico , Proteínas Recombinantes/metabolismo , Termodinâmica , Veias Umbilicais
17.
Mol Biol Cell ; 15(9): 4148-65, 2004 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-15215314

RESUMO

CD147, a type I integral membrane protein of the immunoglobulin superfamily, exhibits reversed polarity in retinal pigment epithelium (RPE). CD147 is apical in RPE in contrast to its basolateral localization in extraocular epithelia. This elicited our interest in understanding the basolateral sorting signals of CD147 in prototypic Madin-Darby canine kidney (MDCK) cells. The cytoplasmic domain of CD147 has basolateral sorting information but is devoid of well-characterized basolateral signals, such as tyrosine and di-leucine motifs. Hence, we carried out systematic site-directed mutagenesis to delineate basolateral targeting information in CD147. Our detailed analysis identified a single leucine (252) as the basolateral targeting motif in the cytoplasmic tail of CD147. Four amino acids (243-246) N-terminal to leucine 252 are also critical basolateral determinants of CD147, because deletion of these amino acids leads to mistargeting of CD147 to the apical membranes. We ruled out the involvement of adaptor complex 1B (AP1B) in the basolateral trafficking of CD147, because LLC-PK1 cells lacking AP1B, target CD147 basolaterally. At variance with MDCK cells, the human RPE cell line ARPE-19 does not distinguish between CD147 (WT) and CD147 with leucine 252 mutated to alanine and targets both proteins apically. Thus, our study identifies an atypical basolateral motif of CD147, which comprises a single leucine and is not recognized by RPE cells. This unusual basolateral sorting signal will be useful in unraveling the specialized sorting machinery of RPE cells.


Assuntos
Antígenos CD/química , Antígenos CD/metabolismo , Epitélio Pigmentado Ocular/metabolismo , Complexo 1 de Proteínas Adaptadoras/metabolismo , Subunidades beta do Complexo de Proteínas Adaptadoras/metabolismo , Sequência de Aminoácidos , Animais , Antígenos CD/genética , Basigina , Linhagem Celular , Polaridade Celular , Sequência Conservada , Cães , Células Epiteliais/citologia , Células Epiteliais/metabolismo , Meia-Vida , Humanos , Células LLC-PK1 , Leucina/química , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Epitélio Pigmentado Ocular/citologia , Sinais Direcionadores de Proteínas/genética , Ratos , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Homologia de Sequência de Aminoácidos , Especificidade da Espécie , Suínos
18.
Methods ; 30(3): 191-7, 2003 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-12798133

RESUMO

SNARE proteins control the membrane fusion events of membrane trafficking pathways. Work in epithelial cells has shown that polarized trafficking to the apical and basolateral plasma membrane domains requires different sets of SNAREs, suggesting a mechanism that contributes to the overall specificity of polarized trafficking and, perhaps, the formation and maintenance of polarity itself. This article describes methods that have been designed and adapted specifically for the investigation of SNAREs in epithelial cells. The knowledge of the subcellular localization of a SNARE of interest is essential to understand its function. Unfortunately, the endogenous expression levels of SNAREs are often low which makes detection challenging. We provide guidelines for determination of the localization of SNAREs by immunofluorescence microscopy including methods for signal amplification, antigen retrieval, and suppression of antibody cross-reactivity. To define which trafficking pathway a SNARE of interest is involved in, one needs to specifically inhibit its function. We provide guidelines for SNARE inhibition by overexpression of the SNARE of interest. An alternative is to introduce inhibitors of SNARE function, such as antibodies or clostridial toxins, into cells. Two methods are presented to make this possible. The first allows the monitoring of effects on trafficking pathways by biochemical assays, and is based on plasma membrane permeabilization using the bacterial toxin streptolysin-O. The second is suitable for single-cell observations and is based on microinjection.


Assuntos
Células Epiteliais/fisiologia , Fusão de Membrana/fisiologia , Proteínas de Membrana/fisiologia , Transporte Proteico/fisiologia , Proteínas de Transporte Vesicular , Proteínas SNARE
19.
Nat Cell Biol ; 5(2): 126-36, 2003 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-12545172

RESUMO

Targeted delivery of proteins to distinct plasma membrane domains is critical to the development and maintenance of polarity in epithelial cells. We used confocal and time-lapse total internal reflection fluorescence microscopy (TIR-FM) to study changes in localization and exocytic sites of post-Golgi transport intermediates (PGTIs) carrying GFP-tagged apical or basolateral membrane proteins during epithelial polarization. In non-polarized Madin Darby Canine Kidney (MDCK) cells, apical and basolateral PGTIs were present throughout the cytoplasm and were observed to fuse with the basal domain of the plasma membrane. During polarization, apical and basolateral PGTIs were restricted to different regions of the cytoplasm and their fusion with the basal membrane was completely abrogated. Quantitative analysis suggested that basolateral, but not apical, PGTIs fused with the lateral membrane in polarized cells, correlating with the restricted localization of Syntaxins 4 and 3 to lateral and apical membrane domains, respectively. Microtubule disruption induced Syntaxin 3 depolarization and fusion of apical PGTIs with the basal membrane, but affected neither the lateral localization of Syntaxin 4 or Sec6, nor promoted fusion of basolateral PGTIs with the basal membrane.


Assuntos
Membrana Celular/metabolismo , Células Epiteliais/metabolismo , Exocitose/fisiologia , Complexo de Golgi/metabolismo , Transporte Proteico , Animais , Anticorpos/metabolismo , Antineoplásicos/farmacologia , Proteínas de Transporte/metabolismo , Linhagem Celular , Polaridade Celular , Cães , Células Epiteliais/citologia , Células Epiteliais/efeitos dos fármacos , Proteínas de Fluorescência Verde , Proteínas Luminescentes/metabolismo , Fusão de Membrana/fisiologia , Proteínas de Membrana/metabolismo , Microinjeções , Microscopia Confocal , Microscopia de Fluorescência , Microtúbulos/metabolismo , Nocodazol/farmacologia , Proteínas Qa-SNARE , Proteínas Recombinantes de Fusão/metabolismo , Fatores de Tempo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA