Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
J Cell Sci ; 133(15)2020 08 04.
Artigo em Inglês | MEDLINE | ID: mdl-32661088

RESUMO

Melanosomes are motile, light-absorbing organelles that are present in pigment cells of the skin and eye. It has been proposed that melanosome localization, in both skin melanocytes and the retinal pigment epithelium (RPE), involves melanosome capture from microtubule motors by an unconventional myosin, which dynamically tethers the melanosomes to actin filaments. Recent studies with melanocytes have questioned this cooperative capture model. Here, we test the model in RPE cells by imaging melanosomes associated with labeled actin filaments and microtubules, and by investigating the roles of different motor proteins. We found that a deficiency in cytoplasmic dynein phenocopies the lack of myosin-7a, in that melanosomes undergo fewer of the slow myosin-7a-dependent movements and are absent from the RPE apical domain. These results indicate that microtubule-based motility is required for the delivery of melanosomes to the actin-rich apical domain and support a capture mechanism that involves both microtubule and actin motors.


Assuntos
Actinas , Melanossomas , Microtúbulos , Miosinas , Epitélio Pigmentado da Retina
2.
Stem Cell Res Ther ; 8(1): 217, 2017 10 02.
Artigo em Inglês | MEDLINE | ID: mdl-28969679

RESUMO

BACKGROUND: Dysfunction of the retinal pigment epithelium (RPE) is implicated in numerous forms of retinal degeneration. The readily accessible environment of the eye makes it particularly suitable for the transplantation of RPE cells, which can now be derived from autologous induced pluripotent stem cells (iPSCs), to treat retinal degeneration. For RPE transplantation to become feasible in the clinic, patient-specific somatic cells should be reprogrammed to iPSCs without the introduction of reprogramming genes into the genome of the host cell, and then subsequently differentiated into RPE cells that are well characterized for safety and functionality prior to transplantation. METHODS: We have reprogrammed human dermal fibroblasts to iPSCs using nonintegrating RNA, and differentiated the iPSCs toward an RPE fate (iPSC-RPE), under Good Manufacturing Practice (GMP)-compatible conditions. RESULTS: Using highly sensitive assays for cell polarity, structure, organelle trafficking, and function, we found that iPSC-RPE cells in culture exhibited key characteristics of native RPE. Importantly, we demonstrate for the first time with any stem cell-derived RPE cell that live cells are able to support dynamic organelle transport. This highly sensitive test is critical for RPE cells intended for transplantation, since defects in intracellular motility have been shown to promote RPE pathogenesis akin to that found in macular degeneration. To test their capabilities for in-vivo transplantation, we injected the iPSC-RPE cells into the subretinal space of a mouse model of retinal degeneration, and demonstrated that the transplanted cells are capable of rescuing lost RPE function. CONCLUSIONS: This report documents the successful generation, under GMP-compatible conditions, of human iPSC-RPE cells that possess specific characteristics of healthy RPE. The report adds to a growing literature on the utility of human iPSC-RPE cells for cell culture investigations on pathogenicity and for therapeutic transplantation, by corroborating findings of others, and providing important new information on essential RPE cell biological properties.


Assuntos
Reprogramação Celular/genética , Vírus da Encefalite Equina Venezuelana/genética , Células Epiteliais/efeitos dos fármacos , Fibroblastos/fisiologia , Células-Tronco Pluripotentes Induzidas/fisiologia , Degeneração Retiniana/terapia , Animais , Diferenciação Celular/efeitos dos fármacos , Polaridade Celular/efeitos dos fármacos , Modelos Animais de Doenças , Vírus da Encefalite Equina Venezuelana/metabolismo , Células Epiteliais/citologia , Células Epiteliais/fisiologia , Células Epiteliais/transplante , Fibroblastos/citologia , Vetores Genéticos/química , Vetores Genéticos/metabolismo , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Pluripotentes Induzidas/efeitos dos fármacos , Injeções Intraoculares , Peptídeos e Proteínas de Sinalização Intercelular/farmacologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Cultura Primária de Células , Degeneração Retiniana/patologia , Degeneração Retiniana/fisiopatologia , Epitélio Pigmentado da Retina/citologia , Epitélio Pigmentado da Retina/efeitos dos fármacos , Epitélio Pigmentado da Retina/fisiologia , Pele/citologia
3.
Nat Biotechnol ; 35(8): 765-772, 2017 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-28504668

RESUMO

Polymorphisms in the human leukocyte antigen (HLA) class I genes can cause the rejection of pluripotent stem cell (PSC)-derived products in allogeneic recipients. Disruption of the Beta-2 Microglobulin (B2M) gene eliminates surface expression of all class I molecules, but leaves the cells vulnerable to lysis by natural killer (NK) cells. Here we show that this 'missing-self' response can be prevented by forced expression of minimally polymorphic HLA-E molecules. We use adeno-associated virus (AAV)-mediated gene editing to knock in HLA-E genes at the B2M locus in human PSCs in a manner that confers inducible, regulated, surface expression of HLA-E single-chain dimers (fused to B2M) or trimers (fused to B2M and a peptide antigen), without surface expression of HLA-A, B or C. These HLA-engineered PSCs and their differentiated derivatives are not recognized as allogeneic by CD8+ T cells, do not bind anti-HLA antibodies and are resistant to NK-mediated lysis. Our approach provides a potential source of universal donor cells for applications where the differentiated derivatives lack HLA class II expression.


Assuntos
Antígenos HLA/imunologia , Células Matadoras Naturais/imunologia , Células-Tronco Pluripotentes/imunologia , Transplantes/imunologia , Animais , Feminino , Rejeição de Enxerto/imunologia , Antígenos HLA/química , Antígenos HLA/genética , Humanos , Camundongos , Células-Tronco Pluripotentes/química , Células-Tronco Pluripotentes/citologia , Transplantes/química , Transplantes/citologia
4.
J Cell Biol ; 210(4): 595-611, 2015 Aug 17.
Artigo em Inglês | MEDLINE | ID: mdl-26261180

RESUMO

The degradation of phagosomes, derived from the ingestion of photoreceptor outer segment (POS) disk membranes, is a major role of the retinal pigment epithelium (RPE). Here, POS phagosomes were observed to associate with myosin-7a, and then kinesin-1, as they moved from the apical region of the RPE. Live-cell imaging showed that the phagosomes moved bidirectionally along microtubules in RPE cells, with kinesin-1 light chain 1 (KLC1) remaining associated in both directions and during pauses. Lack of KLC1 did not inhibit phagosome speed, but run length was decreased, and phagosome localization and degradation were impaired. In old mice, lack of KLC1 resulted in RPE pathogenesis that was strikingly comparable to aspects of age-related macular degeneration (AMD), with an excessive accumulation of RPE and sub-RPE deposits, as well as oxidative and inflammatory stress responses. These results elucidate mechanisms of POS phagosome transport in relation to degradation, and demonstrate that defective microtubule motor transport in the RPE leads to phenotypes associated with AMD.


Assuntos
Degeneração Macular/metabolismo , Proteínas Associadas aos Microtúbulos/genética , Fagossomos/metabolismo , Epitélio Pigmentado da Retina/metabolismo , Animais , Transporte Biológico , Células Cultivadas , Ativação do Complemento , Cinesinas , Degeneração Macular/genética , Degeneração Macular/patologia , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas Associadas aos Microtúbulos/metabolismo , Miosina VIIa , Miosinas/metabolismo , Estresse Oxidativo , Segmento Externo das Células Fotorreceptoras da Retina/metabolismo , Segmento Externo das Células Fotorreceptoras da Retina/patologia
5.
Artigo em Inglês | MEDLINE | ID: mdl-25605753

RESUMO

Usher syndrome is a deaf-blindness disorder. One of the subtypes, Usher 1B, is caused by loss of function of the gene encoding the unconventional myosin, MYO7A. A variety of different viral-based delivery approaches have been tested for retinal gene therapy to prevent the blindness of Usher 1B, and a clinical trial based on one of these approaches has begun. This review evaluates the different approaches.


Assuntos
Terapia Genética/métodos , Mutação/genética , Miosinas/genética , Síndromes de Usher/terapia , Adenoviridae , Animais , Modelos Animais de Doenças , Técnicas de Transferência de Genes , Humanos , Lentivirus , Camundongos , Miosina VIIa , Fenótipo , Síndromes de Usher/genética
6.
Mol Neurobiol ; 52(3): 1135-1151, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25301234

RESUMO

A main requisite in the phagocytosis of ingested material is a coordinated series of maturation steps which lead to the degradation of ingested cargo. Photoreceptor outer segment (POS) renewal involves phagocytosis of the distal disk membranes by the retinal pigment epithelium (RPE). Previously, we identified melanoregulin (MREG) as an intracellular cargo-sorting protein required for the degradation of POS disks. Here, we provide evidence that MREG-dependent processing links both autophagic and phagocytic processes in LC3-associated phagocytosis (LAP). Ingested POS phagosomes are associated with endogenous LC3 and MREG. The LC3 association with POSs exhibited properties of LAP; it was independent of rapamycin pretreatment, but dependent on Atg5. Loss of MREG resulted in a decrease in the extent of LC3-POS association. Studies using DQ-BSA suggest that loss of MREG does not compromise the association and fusion of LC3-positive phagosomes with lysosomes. Furthermore, the mechanism of MREG action is likely through a protein complex that includes LC3, as determined by colocalization and immunoprecipitation in both RPE cells and macrophages. We posit that MREG participates in coordinating the association of phagosomes with LC3 for content degradation with the loss of MREG leading to phagosome accumulation.


Assuntos
Proteínas de Transporte/fisiologia , Proteínas do Olho/fisiologia , Proteínas Associadas aos Microtúbulos/fisiologia , Fagocitose , Fagossomos/metabolismo , Epitélio Pigmentado da Retina/fisiologia , Proteínas Adaptadoras de Transporte Vesicular , Animais , Autofagia/efeitos dos fármacos , Autofagia/fisiologia , Proteína 5 Relacionada à Autofagia , Proteínas de Transporte/genética , Bovinos , Ritmo Circadiano , Humanos , Peptídeos e Proteínas de Sinalização Intracelular , Macrófagos/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Complexos Multiproteicos , Fagocitose/efeitos dos fármacos , Fagocitose/fisiologia , Transporte Proteico , Proteólise , Segmento Externo das Células Fotorreceptoras da Retina/fisiologia , Epitélio Pigmentado da Retina/citologia , Epitélio Pigmentado da Retina/embriologia , Sirolimo/farmacologia
7.
Adv Exp Med Biol ; 801: 725-31, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24664764

RESUMO

Usher syndrome type 1B, which is characterized by congenital deafness and progressive retinal degeneration, is caused by the loss of the function of MYO7A. Prevention of the retinal degeneration should be possible by delivering functional MYO7A to retinal cells. Although this approach has been used successfully in clinical trials for Leber congenital amaurosis (LCA2), it remains a challenge for Usher 1B because of the large size of the MYO7A cDNA. Different viral vectors have been tested for use in MYO7A gene therapy. Here, we review approaches with lentiviruses, which can accommodate larger genes, as well as attempts to use adeno-associated virus (AAV), which has a smaller packaging capacity. In conclusion, both types of viral vector appear to be effective. Despite concerns about the ability of lentiviruses to access the photoreceptor cells, a phenotype of the photoreceptors of Myo7a-mutant mice can be corrected. And although MYO7A cDNA is significantly larger than the nominal carrying capacity of AAV, AAV-MYO7A in single vectors also corrected Myo7a-mutant phenotypes in photoreceptor and RPE cells. Interestingly, however, a dual AAV vector approach was found to be much less effective.


Assuntos
Dependovirus/genética , Terapia Genética/métodos , Lentivirus/genética , Miosinas/genética , Síndromes de Usher/terapia , Animais , Vetores Genéticos , Humanos , Camundongos , Camundongos Knockout , Miosina VIIa , Síndromes de Usher/genética
8.
Tissue Eng Part C Methods ; 20(5): 393-400, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24047104

RESUMO

The current standard method to culture human limbal stem/progenitor cells (LSCs) in vitro is to culture limbal epithelial cells directly on a layer of murine 3T3 feeder cells (standard method). The direct contact between human cells and murine feeder cells poses the potential risk of incomplete removal of feeder cells after culture and cross-contamination in clinical applications. We present here a novel three-dimensional (3D) sandwich method in which LSCs and feeder cells were separately cultured on opposite sides of a porous membrane. Limbal epithelial cells in the form of single-cell suspensions, cell clusters, and tissue explants were subjected to standard culture or to a 3D sandwich culture method. The 3D sandwich method consistently yielded LSCs derived from cell clusters and tissue explants. The expanded LSCs exhibited a small, compact, cuboidal stem-cell morphology and stem cell phenotypes comparable to those of LSCs derived from the standard culture method. Limbal epithelial cell clusters cultured with the sandwich method had a significantly higher proliferation rate than did those cultured with the standard method. The 3D sandwich method did not favor the propagation of single LSCs. In summary, the 3D sandwich method permits complete separation between cultured cells and feeder cells, while providing an even and maximal proximity between them. This alternative method permits culturing of LSCs without the risk of feeder cell contamination.


Assuntos
Técnicas de Cultura de Células/métodos , Limbo da Córnea/citologia , Células-Tronco/citologia , Células 3T3 , Adulto , Idoso , Animais , Agregação Celular , Comunicação Celular , Proliferação de Células , Células Cultivadas , Células Alimentadoras/citologia , Humanos , Queratina-12/metabolismo , Queratina-14/metabolismo , Camundongos , Pessoa de Meia-Idade , Padrões de Referência , Células-Tronco/metabolismo , Suspensões , Fatores de Transcrição/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Adulto Jovem
10.
Biochem Soc Trans ; 39(5): 1207-10, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21936790

RESUMO

Mutations in MYO7A (myosin VIIa) cause Usher syndrome type 1B, a disorder involving profound congenital deafness and progressive blindness. In the retina, most MYO7A is localized in the apical region of the RPE (retinal pigmented epithelial) cells, and a small amount is associated with the ciliary and periciliary membranes of the photoreceptor cells. Its roles appear to be quite varied. Studies with MYO7A-null mice indicate that MYO7A participates in the apical localization of RPE melanosomes and in the removal of phagosomes from the apical RPE for their delivery to lysosomes in the basal RPE. In the first role, MYO7A competes with microtubule motors, but in the second one, it may function co-operatively. An additional role of MYO7A in the RPE is indicated by the requirement for it in the light-dependent translocation of the ER (endoplasmic reticulum)-associated visual cycle enzyme RPE65 and normal functioning of the visual retinoid cycle. In photoreceptor cells lacking MYO7A, opsin accumulates abnormally in the transition zone of the cilium, suggesting that MYO7A functions as a selective barrier for membrane proteins at the distal end of the transition zone. It is likely that the progressive retinal degeneration that occurs in Usher syndrome 1B patients results from a combination of cellular defects in the RPE and photoreceptor cells.


Assuntos
Miosinas/metabolismo , Retina/metabolismo , Síndromes de Usher/metabolismo , Animais , Humanos , Melanossomas/metabolismo , Camundongos , Camundongos Knockout , Mutação , Miosina VIIa , Miosinas/genética , Células Fotorreceptoras/citologia , Células Fotorreceptoras/metabolismo , Células Fotorreceptoras/patologia , Retina/citologia , Retina/patologia , Epitélio Pigmentado da Retina/citologia , Epitélio Pigmentado da Retina/metabolismo , Epitélio Pigmentado da Retina/patologia , Síndromes de Usher/genética , Síndromes de Usher/patologia
11.
Hum Mol Genet ; 20(13): 2560-70, 2011 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-21493626

RESUMO

Mutations in the MYO7A gene cause a deaf-blindness disorder, known as Usher syndrome 1B.  In the retina, the majority of MYO7A is in the retinal pigmented epithelium (RPE), where many of the reactions of the visual retinoid cycle take place.  We have observed that the retinas of Myo7a-mutant mice are resistant to acute light damage. In exploring the basis of this resistance, we found that Myo7a-mutant mice have lower levels of RPE65, the RPE isomerase that has a key role in the retinoid cycle.  We show for the first time that RPE65 normally undergoes a light-dependent translocation to become more concentrated in the central region of the RPE cells.  This translocation requires MYO7A, so that, in Myo7a-mutant mice, RPE65 is partly mislocalized in the light.  RPE65 is degraded more quickly in Myo7a-mutant mice, perhaps due to its mislocalization, providing a plausible explanation for its lower levels.  Following a 50-60% photobleach, Myo7a-mutant retinas exhibited increased all-trans-retinyl ester levels during the initial stages of dark recovery, consistent with a deficiency in RPE65 activity.  Lastly, MYO7A and RPE65 were co-immunoprecipitated from RPE cell lysate by antibodies against either of the proteins, and the two proteins were partly colocalized, suggesting a direct or indirect interaction.  Together, the results support a role for MYO7A in the translocation of RPE65, illustrating the involvement of a molecular motor in the spatiotemporal organization of the retinoid cycle in vision.


Assuntos
Proteínas do Olho/metabolismo , Miosinas/metabolismo , Animais , Linhagem Celular , Modelos Animais de Doenças , Proteínas do Olho/genética , Humanos , Espaço Intracelular/metabolismo , Luz/efeitos adversos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Miosina VIIa , Miosinas/genética , Ligação Proteica/fisiologia , Transporte Proteico/genética , Transporte Proteico/efeitos da radiação , Retina/metabolismo , Retina/patologia , Retina/efeitos da radiação , Degeneração Retiniana/genética , Degeneração Retiniana/patologia , Síndromes de Usher/genética , Síndromes de Usher/metabolismo , Síndromes de Usher/patologia
12.
Nat Genet ; 42(2): 175-80, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-20081859

RESUMO

Degeneration of photoreceptors is a common feature of ciliopathies, owing to the importance of the specialized ciliary structure of these cells. Mutations in AHI1, which encodes a cilium-localized protein, have been shown to cause a form of Joubert syndrome that is highly penetrant for retinal degeneration. We show that Ahi1-null mice fail to form retinal outer segments and have abnormal distribution of opsin throughout their photoreceptors. Apoptotic cell death of photoreceptors occurs rapidly between 2 and 4 weeks of age in these mice and is significantly (P = 0.00175 and 0.00613) delayed by a reduced dosage of opsin. This phenotype also shows dosage-sensitive genetic interactions with Nphp1, another ciliopathy-related gene. Although it is not a primary cause of retinal blindness in humans, we show that an allele of AHI1 is associated with a more than sevenfold increase in relative risk of retinal degeneration within a cohort of individuals with the hereditary kidney disease nephronophthisis. Our data support context-specific roles for AHI1 as a contributor to retinopathy and show that AHI1 may explain a proportion of the variability in retinal phenotypes observed in nephronophthisis.


Assuntos
Células Fotorreceptoras de Vertebrados/patologia , Proteínas Proto-Oncogênicas/metabolismo , Degeneração Retiniana/patologia , Proteínas Adaptadoras de Transdução de Sinal , Proteínas Adaptadoras de Transporte Vesicular , Substituição de Aminoácidos/genética , Animais , Proteínas de Transporte/genética , Morte Celular , Linhagem Celular , Proteínas do Citoesqueleto , Humanos , Itália , Camundongos , Opsinas/metabolismo , Células Fotorreceptoras de Vertebrados/metabolismo , Células Fotorreceptoras de Vertebrados/ultraestrutura , Proteínas Proto-Oncogênicas/deficiência , Proteínas Proto-Oncogênicas/genética , Degeneração Retiniana/genética
13.
Pigment Cell Melanoma Res ; 22(4): 461-73, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19317802

RESUMO

Myrip is a Rab27a and MyosinVIIa (MyoVIIa) linking protein that may regulate melanosome transport in the retinal pigment epithelium (RPE). Myrip also binds MyosinVa (MyoVa) in vitro however it is unclear whether this interaction is of sufficient affinity to be physiologically relevant. Here, we addressed the questions of whether Myrip interacts with MyoVa in cells and the molecular basis of cellular activation of MyoVa and MyoVIIa by Myrip. To answer these questions we used melanosome transport in skin melanocytes and RPE cells as read-outs of MyoVa and MyoVIIa activity. We found that Myrip recruits and activates MyoVa on skin melanosomes with similar efficiency to the established MyoVa activator Melanophilin (Mlph). Mutagenesis showed that a Myrip-Mlph conserved amphipathic helix (MMAH) is essential for MyoVa interaction while other Myrip regions, including the MyoVa exon F binding domain equivalent, play non-essential roles in this interaction. This suggests that, in contrast to Mlph, Myrip interacts with MyoVa lacking melanocyte-specific exon F. Parallel studies of RPE melanosome transport reveal that Myrip-specific inserts, but not the MMAH, are essential for MyoVIIa activation. We conclude that Myrip is a versatile Rab27a-associated myosin-activating protein that mediates cellular activation of MyoVa and MyoVIIa via non-overlapping domains.


Assuntos
Melanossomas/metabolismo , Cadeias Pesadas de Miosina/metabolismo , Miosina Tipo V/metabolismo , Miosinas/metabolismo , Proteínas de Transporte Vesicular/fisiologia , Animais , Transporte Biológico , Linhagem Celular , Feminino , Melanócitos/metabolismo , Camundongos , Camundongos Mutantes , Cadeias Pesadas de Miosina/genética , Miosina Tipo V/genética , Miosina VIIa , Miosinas/genética , Ligação Proteica , Estrutura Terciária de Proteína , Pele/metabolismo , Proteínas de Transporte Vesicular/genética
14.
J Biol Chem ; 284(16): 10877-89, 2009 Apr 17.
Artigo em Inglês | MEDLINE | ID: mdl-19240024

RESUMO

Melanoregulin (MREG), the product of the Mreg(dsu) gene, is a small highly charged protein, hypothesized to play a role in organelle biogenesis due to its effect on pigmentation in dilute, ashen, and leaden mutant mice. Here we provide evidence that MREG is required in lysosome-dependent phagosome degradation. In the Mreg(-/-) mouse, we show that loss of MREG function results in phagosome accumulation due to delayed degradation of engulfed material. Over time, the Mreg(-/-) mouse retinal pigment epithelial cells accumulate the lipofuscin component, A2E. MREG-deficient human and mouse retinal pigment epithelial cells exhibit diminished activity of the lysosomal hydrolase, cathepsin D, due to defective processing. Moreover, MREG localizes to small intracellular vesicles and associates with the endosomal phosphoinositide, phosphatidylinositol 3,5-biphosphate. Collectively, these studies suggest that MREG is required for lysosome maturation and support a role for MREG in intracellular trafficking.


Assuntos
Proteínas de Transporte/metabolismo , Células Epiteliais/metabolismo , Lisossomos/metabolismo , Epitélio Pigmentado Ocular/citologia , Proteínas Adaptadoras de Transporte Vesicular , Animais , Proteínas de Transporte/genética , Catepsina D/metabolismo , Linhagem Celular , Células Epiteliais/citologia , Humanos , Peptídeos e Proteínas de Sinalização Intracelular , Lipofuscina/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fagocitose/fisiologia , Fosfatidiletanolaminas/metabolismo , Fosfatos de Fosfatidilinositol/metabolismo , Compostos de Piridínio/metabolismo , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Retinaldeído/análogos & derivados , Retinaldeído/metabolismo , Retinoides/metabolismo
15.
Traffic ; 8(5): 486-99, 2007 May.
Artigo em Inglês | MEDLINE | ID: mdl-17451552

RESUMO

The retinal pigment epithelium (RPE) contains melanosomes similar to those found in the skin melanocytes, which undergo dramatic light-dependent movements in fish and amphibians. In mammals, those movements are more subtle and appear to be regulated by the Rab27a GTPase and the unconventional myosin, Myosin VIIa (MyoVIIa). Here we address the hypothesis that a recently identified Rab27a- and MyoVIIa-interacting protein, Myrip, promotes the formation of a functional tripartite complex. In heterologous cultured cells, all three proteins co-immunoprecipitated following overexpression. Rab27a and Myrip localize to the peripheral membrane of RPE melanosomes as observed by immunofluorescence and immunoelectron microscopy. Melanosome dynamics were studied using live-cell imaging of mouse RPE primary cultures. Wild-type RPE melanosomes exhibited either stationary or slow movement interrupted by bursts of fast movement, with a peripheral directionality trend. Nocodazole treatment led to melanosome paralysis, suggesting that movement requires microtubule motors. Significant and similar alterations in melanosome dynamics were observed when any one of the three components of the complex was missing, as studied in ashen- (Rab27a defective) and shaker-1 (MyoVIIa mutant)-derived RPE cells, and in wild-type RPE cells transduced with adenovirus carrying specific sequences to knockdown Myrip expression. We observed a significant increase in the number of motile melanosomes, exhibiting more frequent and prolonged bursts of fast movement, and inversion of directionality. Similar alterations were observed upon cytochalasin D treatment, suggesting that the Rab27a-Myrip-MyoVIIa complex regulates tethering of melanosomes onto actin filaments, a process that ensures melanosome movement towards the cell periphery.


Assuntos
Dineínas/metabolismo , Melanossomas/metabolismo , Miosinas/metabolismo , Epitélio Pigmentado Ocular/metabolismo , Proteínas de Transporte Vesicular/fisiologia , Proteínas rab de Ligação ao GTP/metabolismo , Actinas/metabolismo , Animais , Transporte Biológico/efeitos dos fármacos , Células COS , Células Cultivadas , Chlorocebus aethiops , Citocalasina D/farmacologia , Dineínas/genética , Células Epiteliais/citologia , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Imunoprecipitação , Melanossomas/química , Melanossomas/ultraestrutura , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Miosina VIIa , Miosinas/genética , Nocodazol/farmacologia , Epitélio Pigmentado Ocular/citologia , Ligação Proteica/efeitos dos fármacos , RNA Antissenso/genética , Transfecção , Proteínas de Transporte Vesicular/análise , Proteínas rab de Ligação ao GTP/análise , Proteínas rab de Ligação ao GTP/genética , Proteínas rab27 de Ligação ao GTP
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA