Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Nat Commun ; 15(1): 371, 2024 Jan 08.
Artículo en Inglés | MEDLINE | ID: mdl-38191531

RESUMEN

Aurora Kinase A (AURKA) promotes cell proliferation and is overexpressed in different types of polycystic kidney disease (PKD). To understand AURKA's role in regulating renal cyst development we conditionally deleted the gene in mouse models of Autosomal Dominant PKD (ADPKD) and Joubert Syndrome, caused by Polycystin 1 (Pkd1) and Inositol polyphosphate-5-phosphatase E (Inpp5e) mutations respectively. We show that while Aurka is dispensable for collecting duct development and homeostasis, its deletion prevents cyst formation in both disease models. Cross-comparison of transcriptional changes implicated AKT signaling in cyst prevention and we show that (i) AURKA and AKT physically interact, (ii) AURKA regulates AKT activity in a kinase-independent manner and (iii) inhibition of AKT can reduce disease severity. AKT activation also regulates Aurka expression, creating a feed-forward loop driving renal cystogenesis. We find that the AURKA kinase inhibitor Alisertib stabilises the AURKA protein, agonizing its cystogenic functions. These studies identify AURKA as a master regulator of renal cyst development in different types of PKD, functioning in-part via AKT.


Asunto(s)
Aurora Quinasa A , Quistes , Enfermedades Renales Poliquísticas , Riñón Poliquístico Autosómico Dominante , Animales , Ratones , Aurora Quinasa A/genética , Monoéster Fosfórico Hidrolasas , Enfermedades Renales Poliquísticas/genética , Enfermedades Renales Poliquísticas/prevención & control , Proteínas Proto-Oncogénicas c-akt/genética
2.
Nat Commun ; 13(1): 7259, 2022 11 25.
Artículo en Inglés | MEDLINE | ID: mdl-36433978

RESUMEN

Time-lapse mechanical properties of stem cell derived cardiac organoids are important biological cues for understanding contraction dynamics of human heart tissues, cardiovascular functions and diseases. However, it remains difficult to directly, instantaneously and accurately characterize such mechanical properties in real-time and in situ because cardiac organoids are topologically complex, three-dimensional soft tissues suspended in biological media, which creates a mismatch in mechanics and topology with state-of-the-art force sensors that are typically rigid, planar and bulky. Here, we present a soft resistive force-sensing diaphragm based on ultrasensitive resistive nanocracked platinum film, which can be integrated into an all-soft culture well via an oxygen plasma-enabled bonding process. We show that a reliable organoid-diaphragm contact can be established by an 'Atomic Force Microscope-like' engaging process. This allows for instantaneous detection of the organoids' minute contractile forces and beating patterns during electrical stimulation, resuscitation, drug dosing, tissue culture, and disease modelling.


Asunto(s)
Diafragma , Organoides , Humanos , Corazón , Tórax , Fenómenos Mecánicos
3.
Mol Pharm ; 19(3): 843-852, 2022 03 07.
Artículo en Inglés | MEDLINE | ID: mdl-35133169

RESUMEN

Folate receptor (FR) overexpression in a wide range of solid tumors provides an opportunity to develop novel, targeted cancer therapeutics. In this study, we investigated whether prebinding the chemotherapeutic methotrexate (MTX) to folate-binding protein (FBP), the soluble form of FR, would enable the protein to serve as a targeted therapeutic vector, enhancing uptake into tumor cells and improving therapeutic efficacy. In an in vivo study, using an FR-overexpressing KB xenograft model in SCID mice, modest improvement in inhibiting tumor growth was observed for the MTX/FBP mixtures as compared to saline control and free MTX. Surprisingly, FBP alone inhibited tumor growth compared to saline control, free MTX, and FBP/MTX. In order to better understand this effect, we investigated the cytotoxicity of micromolar concentrations of FBP in vitro using the KB, HeLa, and A549 cancer cell lines. Our results revealed concentration-dependent apoptosis (24 h; 10-50 µM) in all three cell lines accompanied by a time- and concentration-dependent reduction (6, 12, and 24 h; 10-50 µM) in metabolic activity and compromised cell plasma membrane integrity. This study demonstrates an apoptosis pathway for cytotoxicity of FBP, an endogenous serum protein, in cancer cell lines with widely varying levels of FR expression. Furthermore, in vivo tumor growth suppression for xenograft KB tumors in SCID mice was observed. These studies suggest novel strategies for the elimination of cancer cells employing endogenous, serum transport proteins.


Asunto(s)
Proteínas Portadoras , Ácido Fólico , Animales , Proteínas Portadoras/metabolismo , Receptores de Folato Anclados a GPI , Ácido Fólico/metabolismo , Humanos , Metotrexato/farmacología , Metotrexato/uso terapéutico , Ratones , Ratones SCID
4.
Adv Healthc Mater ; 10(17): e2100577, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34019737

RESUMEN

In spite of advances in electronics and internet technologies, current healthcare remains hospital-centred. Disruptive technologies are required to translate state-of-art wearable devices into next-generation patient-centered diagnosis and therapy. In this review, recent advances in the emerging field of soft wearable materials and devices are summarized. A prerequisite for such future healthcare devices is the need of novel materials to be mechanically compliant, electrically conductive, and biologically compatible. It is begun with an overview of the two viable design strategies reported in the literatures, which is followed by description of state-of-the-art wearable healthcare devices for monitoring physical, electrophysiological, chemical, and biological signals. Self-powered wearable bioenergy devices are also covered and sensing systems, as well as feedback-controlled wearable closed-loop biodiagnostic and therapy systems. Finally, it is concluded with an overall summary and future perspective.


Asunto(s)
Dispositivos Electrónicos Vestibles , Atención a la Salud , Conductividad Eléctrica , Electrónica , Fenómenos Electrofisiológicos , Humanos
5.
Hum Mol Genet ; 29(1): 31-48, 2020 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-31625572

RESUMEN

Polycystic kidney disease (PKD) results in the formation of renal cysts that can impair function leading to renal failure. DNA damage accumulates in renal epithelial cells in PKD, but the molecular mechanisms are unclear and are investigated here. Phosphoinositide 3-kinase (PI3K)/AKT signaling activates mammalian target of rapamycin complex 1 (mTORC1) and hyperactivation of mTORC1 is a common event in PKD; however, mTORC1 inhibitors have yielded disappointing results in clinical trials. Here, we demonstrate AKT and mTORC1 hyperactivation in two representative murine PKD models (renal epithelial-specific Inpp5e knockout and collecting duct-specific Pkd1 deletion) and identify a downstream signaling network that contributes to DNA damage accumulation. Inpp5e- and Pkd1-null renal epithelial cells showed DNA damage including double-stranded DNA breaks associated with increased replication fork numbers, multinucleation and centrosome amplification. mTORC1 activated CAD, which promotes de novo pyrimidine synthesis, to sustain cell proliferation. AKT, but not mTORC1, inhibited the DNA repair/replication fork origin firing regulator TOPBP1, which impacts on DNA damage and cell proliferation. Notably, Inpp5e- and Pkd1-null renal epithelial cell spheroid formation defects were rescued by AKT inhibition. These data reveal that AKT hyperactivation contributes to DNA damage accumulation in multiple forms of PKD and cooperates with mTORC1 to promote cell proliferation. Hyperactivation of AKT may play a causal role in PKD by regulating DNA damage and cell proliferation, independent of mTORC1, and AKT inhibition may be a novel therapeutic approach for PKD.


Asunto(s)
Daño del ADN/fisiología , Enfermedades Renales Poliquísticas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Animales , Proliferación Celular/genética , Proliferación Celular/fisiología , Células Cultivadas , Daño del ADN/genética , Electroforesis en Gel de Poliacrilamida , Immunoblotting , Inmunohistoquímica , Masculino , Diana Mecanicista del Complejo 1 de la Rapamicina/genética , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Ratones , Fosfatidilinositol 3-Quinasas/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Enfermedades Renales Poliquísticas/genética , Proteínas Proto-Oncogénicas c-akt/genética , Transducción de Señal/genética , Transducción de Señal/fisiología
6.
J Biol Chem ; 294(45): 16684-16697, 2019 11 08.
Artículo en Inglés | MEDLINE | ID: mdl-31543504

RESUMEN

Macrophage phagocytosis is required for effective clearance of invading bacteria and other microbes. Coordinated phosphoinositide signaling is critical both for phagocytic particle engulfment and subsequent phagosomal maturation to a degradative organelle. Phosphatidylinositol 3-phosphate (PtdIns(3)P) is a phosphoinositide that is rapidly synthesized and degraded on phagosomal membranes, where it recruits FYVE domain- and PX motif-containing proteins that promote phagosomal maturation. However, the molecular mechanisms that regulate PtdIns(3)P removal from the phagosome have remained unclear. We report here that a myotubularin PtdIns(3)P 3-phosphatase, myotubularin-related protein-4 (MTMR4), regulates macrophage phagocytosis. MTMR4 overexpression reduced and siRNA-mediated Mtmr4 silencing increased levels of cell-surface immunoglobulin receptors (i.e. Fcγ receptors (FcγRs)) on RAW 264.7 macrophages, associated with altered pseudopodal F-actin. Furthermore, MTMR4 negatively regulated the phagocytosis of IgG-opsonized particles, indicating that MTMR4 inhibits FcγR-mediated phagocytosis, and was dynamically recruited to phagosomes of macrophages during phagocytosis. MTMR4 overexpression decreased and Mtmr4-specific siRNA expression increased the duration of PtdIns(3)P on phagosomal membranes. Macrophages treated with Mtmr4-specific siRNA were more resistant to Mycobacterium marinum-induced phagosome arrest, associated with increased maturation of mycobacterial phagosomes, indicating that extended PtdIns(3)P signaling on phagosomes in the Mtmr4-knockdown cells permitted trafficking of phagosomes to acidic late endosomal and lysosomal compartments. In conclusion, our findings indicate that MTMR4 regulates PtdIns(3)P degradation in macrophages and thereby controls phagocytosis and phagosomal maturation.


Asunto(s)
Fagocitosis , Fagosomas/metabolismo , Fosfatos de Fosfatidilinositol/metabolismo , Proteínas Tirosina Fosfatasas no Receptoras/metabolismo , Actinas/metabolismo , Animales , Endosomas/metabolismo , Humanos , Inmunoglobulina G/inmunología , Lisosomas/metabolismo , Macrófagos/citología , Macrófagos/metabolismo , Ratones , Mycobacterium marinum/patogenicidad , Proteínas Tirosina Fosfatasas no Receptoras/antagonistas & inhibidores , Proteínas Tirosina Fosfatasas no Receptoras/genética , Células RAW 264.7 , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Receptores de IgG/metabolismo , Transducción de Señal
7.
Hum Mol Genet ; 28(2): 230-244, 2019 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-30265301

RESUMEN

Polycystic kidney disease (PKD) results from excessive renal epithelial cell proliferation, leading to the formation of large fluid filled cysts which impair renal function and frequently lead to renal failure. Hyperactivation of numerous signaling pathways is hypothesized to promote renal epithelial cell hyperproliferation including mTORC1, extracellular signal-regulated kinase (ERK) and WNT signaling. ß-catenin and its target genes are overexpressed in some PKD models and expression of activated ß-catenin induces cysts in mice; however, ß-catenin murine knockout studies indicate it may also inhibit cystogenesis. Therefore, it remains unclear whether ß-catenin is pro- or anti-cystogenic and whether its role is canonical WNT signaling-dependent. Here, we investigate whether ß-catenin deletion in a PKD model with hyperactived ß-catenin signaling affects disease progression to address whether increased ß-catenin drives PKD. We used renal epithelial cell specific Inpp5e-null PKD mice which we report exhibit increased ß-catenin and target gene expression in the cystic kidneys. Surprisingly, co-deletion of ß-catenin with Inpp5e in renal epithelial cells exacerbated polycystic kidney disease and renal failure compared to Inpp5e deletion alone, but did not normalize ß-catenin target gene expression. ß-catenin/Inpp5e double-knockout kidneys exhibited increased cyst initiation, cell proliferation and MEK/ERK signaling compared to Inpp5e-null, associated with increased fibrosis, which may collectively contribute to accelerated disease. Therefore, increased ß-catenin and WNT target gene expression are not necessarily cyst promoting. Rather ß-catenin may play a dual and context-dependent role in PKD and in the presence of other cyst-inducing mutations (Inpp5e-deletion); ß-catenin loss may exacerbate disease in a WNT target gene-independent manner.


Asunto(s)
Enfermedades Renales Poliquísticas/metabolismo , beta Catenina/metabolismo , Animales , Proliferación Celular , Células Cultivadas , Progresión de la Enfermedad , Eliminación de Gen , Expresión Génica , Riñón/metabolismo , Sistema de Señalización de MAP Quinasas , Ratones , Ratones Noqueados , Monoéster Fosfórico Hidrolasas/genética , Enfermedades Renales Poliquísticas/enzimología , Enfermedades Renales Poliquísticas/genética , Transducción de Señal , Proteínas Wnt/metabolismo , beta Catenina/genética
8.
J Cell Biol ; 216(1): 247-263, 2017 Jan 02.
Artículo en Inglés | MEDLINE | ID: mdl-27998989

RESUMEN

Human ciliopathies, including Joubert syndrome (JBTS), arise from cilia dysfunction. The inositol polyphosphate 5-phosphatase INPP5E localizes to cilia and is mutated in JBTS. Murine Inpp5e ablation is embryonically lethal and recapitulates JBTS, including neural tube defects and polydactyly; however, the underlying defects in cilia signaling and the function of INPP5E at cilia are still emerging. We report Inpp5e-/- embryos exhibit aberrant Hedgehog-dependent patterning with reduced Hedgehog signaling. Using mouse genetics, we show increasing Hedgehog signaling via Smoothened M2 expression rescues some Inpp5e-/- ciliopathy phenotypes and "normalizes" Hedgehog signaling. INPP5E's phosphoinositide substrates PI(4,5)P2 and PI(3,4,5)P3 accumulated at the transition zone (TZ) in Hedgehog-stimulated Inpp5e-/- cells, which was associated with reduced recruitment of TZ scaffolding proteins and reduced Smoothened levels at cilia. Expression of wild-type, but not 5-phosphatase-dead, INPP5E restored TZ molecular organization and Smoothened accumulation at cilia. Therefore, we identify INPP5E as an essential point of convergence between Hedgehog and phosphoinositide signaling at cilia that maintains TZ function and Hedgehog-dependent embryonic development.


Asunto(s)
Anomalías Múltiples/enzimología , Cerebelo/anomalías , Cilios/enzimología , Embrión de Mamíferos/enzimología , Anomalías del Ojo/enzimología , Enfermedades Renales Quísticas/enzimología , Fosfatidilinositol 4,5-Difosfato/metabolismo , Fosfatos de Fosfatidilinositol/metabolismo , Monoéster Fosfórico Hidrolasas/metabolismo , Retina/anomalías , Epitelio Pigmentado de la Retina/enzimología , Sistemas de Mensajero Secundario , Anomalías Múltiples/genética , Animales , Línea Celular , Cerebelo/enzimología , Modelos Animales de Enfermedad , Anomalías del Ojo/genética , Regulación del Desarrollo de la Expresión Génica , Predisposición Genética a la Enfermedad , Proteínas Hedgehog/genética , Proteínas Hedgehog/metabolismo , Humanos , Enfermedades Renales Quísticas/genética , Factores de Transcripción de Tipo Kruppel/genética , Factores de Transcripción de Tipo Kruppel/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Fenotipo , Monoéster Fosfórico Hidrolasas/deficiencia , Monoéster Fosfórico Hidrolasas/genética , Retina/enzimología , Receptor Smoothened/genética , Receptor Smoothened/metabolismo , Factores de Tiempo , Transfección , Proteína Gli2 con Dedos de Zinc
9.
Hum Mol Genet ; 25(11): 2295-2313, 2016 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-27056978

RESUMEN

Polycystic kidney disease (PKD) is a common cause of renal failure with few effective treatments. INPP5E is an inositol polyphosphate 5-phosphatase that dephosphorylates phosphoinositide 3-kinase (PI3K)-generated PI(3,4,5)P3 and is mutated in ciliopathy syndromes. Germline Inpp5e deletion is embryonically lethal, attributed to cilia stability defects, and is associated with polycystic kidneys. However, the molecular mechanisms responsible for PKD development upon Inpp5e loss remain unknown. Here, we show conditional inactivation of Inpp5e in mouse kidney epithelium results in severe PKD and renal failure, associated with a partial reduction in cilia number and hyperactivation of PI3K/Akt and downstream mammalian target of rapamycin complex 1 (mTORC1) signaling. Treatment with an mTORC1 inhibitor improved kidney morphology and function, but did not affect cilia number or length. Therefore, we identify Inpp5e as an essential inhibitor of the PI3K/Akt/mTORC1 signaling axis in renal epithelial cells, and demonstrate a critical role for Inpp5e-dependent mTORC1 regulation in PKD suppression.


Asunto(s)
Riñón/metabolismo , Complejos Multiproteicos/genética , Monoéster Fosfórico Hidrolasas/genética , Enfermedades Renales Poliquísticas/genética , Serina-Treonina Quinasas TOR/genética , Animales , Ciliopatías/tratamiento farmacológico , Ciliopatías/genética , Ciliopatías/patología , Modelos Animales de Enfermedad , Elafina/genética , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Células Epiteliales/patología , Mutación de Línea Germinal , Humanos , Riñón/efectos de los fármacos , Riñón/patología , Diana Mecanicista del Complejo 1 de la Rapamicina , Ratones , Complejos Multiproteicos/antagonistas & inhibidores , Monoéster Fosfórico Hidrolasas/antagonistas & inhibidores , Enfermedades Renales Poliquísticas/tratamiento farmacológico , Enfermedades Renales Poliquísticas/patología , Proteínas Proto-Oncogénicas c-akt/genética , Eliminación de Secuencia , Transducción de Señal/efectos de los fármacos , Sirolimus/administración & dosificación , Serina-Treonina Quinasas TOR/antagonistas & inhibidores
10.
J Cell Sci ; 128(2): 364-72, 2015 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-25395580

RESUMEN

Mutations in inositol polyphosphate 5-phosphatase E (INPP5E) cause the ciliopathies known as Joubert and MORM syndromes; however, the role of INPP5E in ciliary biology is not well understood. Here, we describe an interaction between INPP5E and AURKA, a centrosomal kinase that regulates mitosis and ciliary disassembly, and we show that this interaction is important for the stability of primary cilia. Furthermore, AURKA phosphorylates INPP5E and thereby increases its 5-phosphatase activity, which in turn promotes transcriptional downregulation of AURKA, partly through an AKT-dependent mechanism. These findings establish the first direct link between AURKA and phosphoinositide signaling and suggest that the function of INPP5E in cilia is at least partly mediated by its interactions with AURKA.


Asunto(s)
Aurora Quinasa A/metabolismo , Cilios/metabolismo , Fosfatidilinositoles/metabolismo , Monoéster Fosfórico Hidrolasas/metabolismo , Anomalías Múltiples/genética , Anomalías Múltiples/patología , Aurora Quinasa A/genética , Cerebelo/anomalías , Cerebelo/patología , Cilios/genética , Anomalías del Ojo/genética , Anomalías del Ojo/patología , Regulación de la Expresión Génica , Humanos , Enfermedades Renales Quísticas/genética , Enfermedades Renales Quísticas/patología , Mitosis/genética , Mutación , Monoéster Fosfórico Hidrolasas/genética , Mapas de Interacción de Proteínas/genética , Retina/anomalías , Retina/patología , Transducción de Señal
11.
Cancer Res ; 73(16): 5066-79, 2013 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-23801747

RESUMEN

It is now clear that progression from localized prostate cancer to incurable castrate-resistant prostate cancer (CRPC) is driven by continued androgen receptor (AR), signaling independently of androgen. Thus, there remains a strong rationale to suppress AR activity as the single most important therapeutic goal in CRPC treatment. Although the expression of ligand-independent AR splice variants confers resistance to AR-targeted therapy and progression to lethal castrate-resistant cancer, the molecular regulators of AR activity in CRPC remain unclear, in particular those pathways that potentiate the function of mutant AR in CRPC. Here, we identify FHL2 as a novel coactivator of ligand-independent AR variants that are important in CRPC. We show that the nuclear localization of FHL2 and coactivation of the AR is driven by calpain cleavage of the cytoskeletal protein filamin, a pathway that shows differential activation in prostate epithelial versus prostate cancer cell lines. We further identify a novel FHL2-AR-filamin transcription complex, revealing how deregulation of this axis promotes the constitutive, ligand-independent activation of AR variants, which are present in CRPC. Critically, the calpain-cleaved filamin fragment and FHL2 are present in the nucleus only in CRPC and not benign prostate tissue or localized prostate cancer. Thus, our work provides mechanistic insight into the enhanced AR activation, most notably of the recently identified AR variants, including AR-V7 that drives CRPC progression. Furthermore, our results identify the first disease-specific mechanism for deregulation of FHL2 nuclear localization during cancer progression. These results offer general import beyond prostate cancer, given that nuclear FHL2 is characteristic of other human cancers where oncogenic transcription factors that drive disease are activated like the AR in prostate cancer.


Asunto(s)
Proteínas con Homeodominio LIM/genética , Proteínas con Homeodominio LIM/metabolismo , Proteínas Musculares/genética , Proteínas Musculares/metabolismo , Neoplasias de la Próstata Resistentes a la Castración/genética , Neoplasias de la Próstata Resistentes a la Castración/metabolismo , Receptores Androgénicos/genética , Receptores Androgénicos/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Animales , Células COS , Calpaína/metabolismo , Línea Celular Tumoral , Núcleo Celular/genética , Núcleo Celular/metabolismo , Chlorocebus aethiops , Células Epiteliales/metabolismo , Células Epiteliales/patología , Filaminas/genética , Filaminas/metabolismo , Humanos , Ligandos , Masculino , Neoplasias de la Próstata Resistentes a la Castración/patología , Activación Transcripcional
12.
FEBS Lett ; 586(18): 2846-57, 2012 Aug 31.
Artículo en Inglés | MEDLINE | ID: mdl-22828281

RESUMEN

Phosphoinositides regulate numerous cellular events via the recruitment and activation of multiple lipid-binding effector proteins. The precise temporal and spatial regulation of phosphoinositide signals by the co-ordinated activities of phosphoinositide kinases and phosphatases is essential for homeostasis and development. Mutations in two inositol polyphosphate 5-phosphatases, INPP5E and OCRL, cause the cerebrorenal syndromes of Joubert and Lowe's, respectively. INPP5E and OCRL exhibit overlapping phosphoinositide substrate specificity and subcellular localisation, including an association with the primary cilia. Here, we review recent studies that identify a new role for these enzymes in the regulation of primary cilia function. Joubert syndrome has been extensively linked to primary cilia defects, and Lowe's may represent a new class of 'ciliopathy associated' syndromes.


Asunto(s)
Cilios/patología , Cilios/fisiología , Monoéster Fosfórico Hidrolasas/metabolismo , Humanos , Inositol Polifosfato 5-Fosfatasas , Mutación , Fosfatidilinositoles/metabolismo , Monoéster Fosfórico Hidrolasas/genética , Síndrome
13.
Dev Biol ; 368(2): 193-202, 2012 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-22609552

RESUMEN

The Hedgehog (HH) signaling pathway is a central regulator of embryonic development, controlling the pattern and proliferation of a wide variety of organs. Previous studies have implicated the secreted protein, Scube2, in HH signal transduction in the zebrafish embryo (Hollway et al., 2006; Kawakami et al., 2005; Woods and Talbot, 2005) although the nature of the molecular function of Scube2 in this process has remained undefined. This analysis has been compounded by the fact that removal of Scube2 activity in the zebrafish embryo leads to only subtle defects in HH signal transduction in vivo (Barresi et al., 2000; Hollway et al., 2006; Ochi and Westerfield, 2007; van Eeden et al., 1996; Wolff et al., 2003). Here we present the discovery of two additional scube genes in zebrafish, scube1 and scube3, and demonstrate their roles in facilitating HH signal transduction. Knocking down the function of all three scube genes simultaneously phenocopies a complete loss of HH signal transduction in the embryo, revealing that Scube signaling is essential for HH signal transduction in vivo. We further define the molecular role of scube2 in HH signaling.


Asunto(s)
Proteínas de Unión al Calcio/genética , Embrión no Mamífero/metabolismo , Proteínas de la Matriz Extracelular/genética , Proteínas Hedgehog/genética , Transducción de Señal/genética , Proteínas de Pez Cebra/genética , Animales , Western Blotting , Células COS , Proteínas de Unión al Calcio/metabolismo , Chlorocebus aethiops , ADN Complementario/química , ADN Complementario/genética , Embrión no Mamífero/embriología , Proteínas de la Matriz Extracelular/metabolismo , Perfilación de la Expresión Génica , Regulación del Desarrollo de la Expresión Génica , Técnicas de Silenciamiento del Gen , Proteínas Hedgehog/metabolismo , Hibridación in Situ , Datos de Secuencia Molecular , Familia de Multigenes , Mutación , Fenotipo , Análisis de Secuencia de ADN , Pez Cebra/embriología , Pez Cebra/genética , Pez Cebra/metabolismo , Proteínas de Pez Cebra/metabolismo
14.
Subcell Biochem ; 58: 215-79, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22403078

RESUMEN

Phosphoinositide phosphatases comprise several large enzyme families with over 35 mammalian enzymes identified to date that degrade many phosphoinositide signals. Growth factor or insulin stimulation activates the phosphoinositide 3-kinase that phosphorylates phosphatidylinositol (4,5)-bisphosphate [PtdIns(4,5)P(2)] to form phosphatidylinositol (3,4,5)-trisphosphate [PtdIns(3,4,5)P(3)], which is rapidly dephosphorylated either by PTEN (phosphatase and tensin homologue deleted on chromosome 10) to PtdIns(4,5)P(2), or by the 5-phosphatases (inositol polyphosphate 5-phosphatases), generating PtdIns(3,4)P(2). 5-phosphatases also hydrolyze PtdIns(4,5)P(2) forming PtdIns(4)P. Ten mammalian 5-phosphatases have been identified, which regulate hematopoietic cell proliferation, synaptic vesicle recycling, insulin signaling, and embryonic development. Two 5-phosphatase genes, OCRL and INPP5E are mutated in Lowe and Joubert syndrome respectively. SHIP [SH2 (Src homology 2)-domain inositol phosphatase] 2, and SKIP (skeletal muscle- and kidney-enriched inositol phosphatase) negatively regulate insulin signaling and glucose homeostasis. SHIP2 polymorphisms are associated with a predisposition to insulin resistance. SHIP1 controls hematopoietic cell proliferation and is mutated in some leukemias. The inositol polyphosphate 4-phosphatases, INPP4A and INPP4B degrade PtdIns(3,4)P(2) to PtdIns(3)P and regulate neuroexcitatory cell death, or act as a tumor suppressor in breast cancer respectively. The Sac phosphatases degrade multiple phosphoinositides, such as PtdIns(3)P, PtdIns(4)P, PtdIns(5)P and PtdIns(3,5)P(2) to form PtdIns. Mutation in the Sac phosphatase gene, FIG4, leads to a degenerative neuropathy. Therefore the phosphatases, like the lipid kinases, play major roles in regulating cellular functions and their mutation or altered expression leads to many human diseases.


Asunto(s)
Neoplasias de la Mama/enzimología , Leucemia/enzimología , Síndrome Oculocerebrorrenal/enzimología , Fosfohidrolasa PTEN/metabolismo , Monoéster Fosfórico Hidrolasas/metabolismo , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Diglicéridos/metabolismo , Femenino , Regulación de la Expresión Génica , Humanos , Inositol 1,4,5-Trifosfato/metabolismo , Inositol Polifosfato 5-Fosfatasas , Leucemia/genética , Leucemia/patología , Síndrome Oculocerebrorrenal/genética , Síndrome Oculocerebrorrenal/patología , Fosfohidrolasa PTEN/genética , Fosfatidilinositol 4,5-Difosfato/metabolismo , Fosfatidilinositol-3,4,5-Trifosfato 5-Fosfatasas , Monoéster Fosfórico Hidrolasas/genética , Sistemas de Mensajero Secundario
15.
J Biol Chem ; 286(34): 29758-70, 2011 Aug 26.
Artículo en Inglés | MEDLINE | ID: mdl-21712384

RESUMEN

Phosphoinositide 3-kinase (PI3K) regulates cell polarity and migration by generating phosphatidylinositol 3,4,5-trisphosphate (PI(3,4,5)P(3)) at the leading edge of migrating cells. The serine-threonine protein kinase Akt binds to PI(3,4,5)P(3), resulting in its activation. Active Akt promotes spatially regulated actin cytoskeletal remodeling and thereby directed cell migration. The inositol polyphosphate 5-phosphatases (5-ptases) degrade PI(3,4,5)P(3) to form PI(3,4)P(2), which leads to diminished Akt activation. Several 5-ptases, including SKIP and SHIP2, inhibit actin cytoskeletal reorganization by opposing PI3K/Akt signaling. In this current study, we identify a molecular co-chaperone termed silencer of death domains (SODD/BAG4) that forms a complex with several 5-ptase family members, including SKIP, SHIP1, and SHIP2. The interaction between SODD and SKIP exerts an inhibitory effect on SKIP PI(3,4,5)P(3) 5-ptase catalytic activity and consequently enhances the recruitment of PI(3,4,5)P(3)-effectors to the plasma membrane. In contrast, SODD(-/-) mouse embryonic fibroblasts exhibit reduced Akt-Ser(473) and -Thr(308) phosphorylation following EGF stimulation, associated with increased SKIP PI(3,4,5)P(3)-5-ptase activity. SODD(-/-) mouse embryonic fibroblasts exhibit decreased EGF-stimulated F-actin stress fibers, lamellipodia, and focal adhesion complexity, a phenotype that is rescued by the expression of constitutively active Akt1. Furthermore, reduced cell migration was observed in SODD(-/-) macrophages, which express the three 5-ptases shown to interact with SODD (SKIP, SHIP1, and SHIP2). Therefore, this study identifies SODD as a novel regulator of PI3K/Akt signaling to the actin cytoskeleton.


Asunto(s)
Actinas/metabolismo , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Monoéster Fosfórico Hidrolasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal/fisiología , Fibras de Estrés/metabolismo , Actinas/genética , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Movimiento Celular/fisiología , Embrión de Mamíferos/metabolismo , Fibroblastos/metabolismo , Adhesiones Focales/genética , Adhesiones Focales/metabolismo , Inositol Polifosfato 5-Fosfatasas , Ratones , Ratones Noqueados , Fosfatidilinositol 3-Quinasas/genética , Fosfatidilinositol-3,4,5-Trifosfato 5-Fosfatasas , Monoéster Fosfórico Hidrolasas/antagonistas & inhibidores , Monoéster Fosfórico Hidrolasas/genética , Proteínas Proto-Oncogénicas c-akt/genética , Seudópodos/genética , Seudópodos/metabolismo , Fibras de Estrés/genética
16.
J Biol Chem ; 286(26): 23407-18, 2011 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-21550974

RESUMEN

Neuron polarization is essential for the formation of one axon and multiple dendrites, establishing the neuronal circuitry. Phosphoinositide 3-kinase (PI3K) signaling promotes axon selection and elongation. Here we report in hippocampal neurons siRNA knockdown of the proline-rich inositol polyphosphate 5-phosphatase (PIPP), which degrades PI3K-generated PtdIns(3,4,5)P(3), results in multiple hyperelongated axons consistent with a polarization defect. We identify collapsin response mediator protein 2 (CRMP2), which regulates axon selection by promoting WAVE1 delivery via Kinesin-1 motors to the axon growth cone, as a PIPP-interacting protein by Y2H screening, direct binding studies, and coimmunoprecipitation of an endogenous PIPP, CRMP2, and Kinesin-1 complex from brain lysates. The C-terminal growth cone-targeting domain of PIPP facilitates its interaction with CRMP2. PIPP growth cone localization is CRMP2-dependent. PIPP knockdown in PC12 cells promotes neurite elongation, WAVE1 and Kinesin-1 growth cone localization, whereas knockdown of CRMP2 exhibits the opposite phenotype, with shorter neurites and decreased WAVE1/Kinesin-1 at the growth cone. In contrast, CRMP2 overexpression promotes neurite elongation, a phenotype rescued by full-length PIPP, or expression of the CRMP2-binding PIPP domain. Therefore this study identifies PIPP and CRMP2 exert opposing roles in promoting axon selection and neurite elongation and the complex between these proteins serves to regulate the localization of effectors that promote neurite extension.


Asunto(s)
Conos de Crecimiento/metabolismo , Hipocampo/metabolismo , Complejos Multiproteicos/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Neuritas/metabolismo , Monoéster Fosfórico Hidrolasas/metabolismo , Animales , Hipocampo/citología , Inositol Polifosfato 5-Fosfatasas , Péptidos y Proteínas de Señalización Intercelular , Cinesinas/genética , Cinesinas/metabolismo , Masculino , Complejos Multiproteicos/genética , Proteínas del Tejido Nervioso/genética , Células PC12 , Fosfatidilinositol 3-Quinasas/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Fosfatos de Fosfatidilinositol/genética , Fosfatos de Fosfatidilinositol/metabolismo , Monoéster Fosfórico Hidrolasas/genética , Ratas , Ratas Sprague-Dawley , Familia de Proteínas del Síndrome de Wiskott-Aldrich/genética , Familia de Proteínas del Síndrome de Wiskott-Aldrich/metabolismo
17.
J Cell Sci ; 123(Pt 18): 3071-83, 2010 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-20736309

RESUMEN

Phosphatidylinositol 3-phosphate [PtdIns(3)P] regulates endocytic trafficking and the sorting of receptors through early endosomes, including the rapid recycling of transferrin (Tfn). However, the phosphoinositide phosphatase that selectively opposes this function is unknown. The myotubularins are a family of eight catalytically active and six inactive enzymes that hydrolyse PtdIns(3)P to form PtdIns. However, the role each myotubularin family member plays in regulating endosomal PtdIns(3)P and thereby endocytic trafficking is not well established. Here, we identify the myotubularin family member MTMR4, which localizes to early endosomes and also to Rab11- and Sec15-positive recycling endosomes. In cells with MTMR4 knockdown, or following expression of the catalytically inactive MTMR4, MTMR4(C407A), the number of PtdIns(3)P-decorated endosomes significantly increased. MTMR4 overexpression delayed the exit of Tfn from early endosomes and its recycling to the plasma membrane. By contrast, expression of MTMR4(C407A), which acts as a dominant-negative construct, significantly accelerated Tfn recycling. However, in MTMR4 knockdown cells Tfn recycling was unchanged, suggesting that other MTMs might also contribute to recycling. MTMR4 regulated the subcellular distribution of Rab11 and, in cells with RNAi-mediated knockdown of MTMR4, Rab11 was directed away from the pericentriolar recycling compartment. The subcellular distribution of VAMP3, a v-SNARE protein that resides in recycling endosomes and endosome-derived transport vesicles, was also regulated by MTMR4. Therefore, MTMR4 localizes at the interface of early and recycling endosomes to regulate trafficking through this pathway.


Asunto(s)
Endosomas/enzimología , Proteínas Tirosina Fosfatasas no Receptoras/metabolismo , Animales , Transporte Biológico , Células COS , Línea Celular , Chlorocebus aethiops , Endosomas/genética , Endosomas/metabolismo , Humanos , Fosfatos de Fosfatidilinositol/metabolismo , Transporte de Proteínas , Proteínas Tirosina Fosfatasas no Receptoras/genética
18.
Methods Mol Biol ; 462: 223-39, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19160673

RESUMEN

Phosphatidylinositol 3,4,5 trisphosphate [PtdIns(3,4,5)P3] is a potent membrane-bound signaling molecule transiently synthesized by phosphoinositide 3-kinase (PI3-kinase) in response to extracellular agonists. PtdIns(3,4,5)P3 signals need to be strictly controlled. PtdIns(3,4,5)P3 recruits and binds effectors that function in oncogenic signaling pathways. PtdIns(3,4,5)P3 activates cell proliferation, growth, and migration as well as regulating insulin signaling. The inositol polyphosphate 5-phosphatase family of enzymes dephosphorylate and thereby modulate PtdIns(3,4,5)P3 levels, attenuating PI3-kinase-dependent signaling. PtdIns(3,4,5)P3 5-phosphatase enzyme activity can be assessed in vitro by analysis of the hydrolysis of radiolabeled or fluorescently labeled PtdIns(3,4,5)P3 and in vivo by visualization of the recruitment and turnover of the PtdIns(3,4,5)P3-specific biosensor GFP-PH/ ARNO or other PtdIns(3,4,5)P3 binding proteins at the plasma membrane.


Asunto(s)
Monoéster Fosfórico Hidrolasas/análisis , Monoéster Fosfórico Hidrolasas/metabolismo , Animales , Western Blotting , Células COS , Chlorocebus aethiops , Técnica del Anticuerpo Fluorescente , Inmunoprecipitación , Microscopía Confocal , Monoéster Fosfórico Hidrolasas/aislamiento & purificación , Proteínas Recombinantes/análisis , Proteínas Recombinantes/aislamiento & purificación , Proteínas Recombinantes/metabolismo , Coloración y Etiquetado , Transfección
19.
J Biol Chem ; 283(33): 22498-504, 2008 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-18577516

RESUMEN

The cytotoxic lymphocyte protease granzyme B (GrB) is elevated in the plasma of individuals with diseases that elicit a cytotoxic lymphocyte-mediated immune response. Given the recently recognized ability of GrB to cleave extracellular matrix proteins, we examined the effect of GrB on the pro-hemostatic molecule von Willebrand factor (VWF). GrB delays ristocetin-induced platelet aggregation and inhibits platelet adhesion and spreading on immobilized VWF under static conditions. It efficiently cleaves VWF at two sites within the A1-3 domains that are essential for the VWF-platelet interaction. Like the VWF regulatory proteinase ADAMTS-13, GrB-mediated cleavage is dependent upon VWF conformation. In vitro, GrB cannot cleave the VWF conformer found in solution, but cleavage is induced when VWF is artificially unfolded or presented as a matrix. GrB cleaves VWF with comparable efficiency to ADAMTS-13 and rapidly processes ultra-large VWF multimers released from activated endothelial cells under physiological shear. GrB also cleaves the matrix form of fibrinogen at several sites. These studies suggest extracellular GrB may help control localized coagulation during inflammation.


Asunto(s)
Granzimas/farmacología , Hemostasis/fisiología , Linfocitos/inmunología , Adhesividad Plaquetaria/efectos de los fármacos , Agregación Plaquetaria/efectos de los fármacos , Factor de von Willebrand/metabolismo , Animales , Endotelio Vascular/fisiología , Fibrinógeno/fisiología , Hemostasis/efectos de los fármacos , Humanos , Cinética , Linfocitos/efectos de los fármacos , Linfocinas/fisiología , Ratones , Conejos
20.
J Biol Chem ; 281(11): 7666-83, 2006 Mar 17.
Artículo en Inglés | MEDLINE | ID: mdl-16407297

RESUMEN

Four and a half LIM protein 1 (FHL1/SLIM1) is highly expressed in skeletal and cardiac muscle; however, the function of FHL1 remains unknown. Yeast two-hybrid screening identified slow type skeletal myosin-binding protein C as an FHL1 binding partner. Myosin-binding protein C is the major myosin-associated protein in striated muscle that enhances the lateral association and stabilization of myosin thick filaments and regulates actomyosin interactions. The interaction between FHL1 and myosin-binding protein C was confirmed using co-immunoprecipitation of recombinant and endogenous proteins. Recombinant FHL2 and FHL3 also bound myosin-binding protein C. FHL1 impaired co-sedimentation of myosin-binding protein C with reconstituted myosin filaments, suggesting FHL1 may compete with myosin for binding to myosin-binding protein C. In intact skeletal muscle and isolated myofibrils, FHL1 localized to the I-band, M-line, and sarcolemma, co-localizing with myosin-binding protein C at the sarcolemma in intact skeletal muscle. Furthermore, in isolated myofibrils FHL1 staining at the M-line appeared to extend partially into the C-zone of the A-band, where it co-localized with myosin-binding protein C. Overexpression of FHL1 in differentiating C2C12 cells induced "sac-like" myotube formation (myosac), associated with impaired Z-line and myosin thick filament assembly. This phenotype was rescued by co-expression of myosin-binding protein C. FHL1 knockdown using RNAi resulted in impaired myosin thick filament formation associated with reduced incorporation of myosin-binding protein C into the sarcomere. This study identified FHL1 as a novel regulator of myosin-binding protein C activity and indicates a role for FHL1 in sarcomere assembly.


Asunto(s)
Proteínas Portadoras/fisiología , Proteínas Musculares/fisiología , Miosinas/química , Sarcómeros/metabolismo , Actomiosina/química , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Células COS , Proteínas Portadoras/metabolismo , Diferenciación Celular , Proliferación Celular , Chlorocebus aethiops , Electroforesis en Gel de Poliacrilamida , Glutatión Transferasa/metabolismo , Humanos , Inmunohistoquímica , Inmunoprecipitación , Péptidos y Proteínas de Señalización Intracelular , Proteínas con Dominio LIM , Ratones , Microscopía Fluorescente , Datos de Secuencia Molecular , Proteínas Musculares/metabolismo , Músculo Esquelético/metabolismo , Miocitos Cardíacos/metabolismo , Oligonucleótidos/química , Péptidos/química , Fenotipo , Unión Proteica , Estructura Terciaria de Proteína , Interferencia de ARN , Proteínas Recombinantes/química , Homología de Secuencia de Aminoácido , Transfección , Técnicas del Sistema de Dos Híbridos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...