Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Proc Natl Acad Sci U S A ; 120(25): e2300310120, 2023 06 20.
Artigo em Inglês | MEDLINE | ID: mdl-37307465

RESUMO

The protein kinase WNK1 (with-no-lysine 1) influences trafficking of ion and small-molecule transporters and other membrane proteins as well as actin polymerization state. We investigated the possibility that actions of WNK1 on both processes are related. Strikingly, we identified the E3 ligase tripartite motif-containing 27 (TRIM27) as a binding partner for WNK1. TRIM27 is involved in fine tuning the WASH (Wiskott-Aldrich syndrome protein and SCAR homologue) regulatory complex which regulates endosomal actin polymerization. Knockdown of WNK1 reduced the formation of the complex between TRIM27 and its deubiquitinating enzyme USP7 (ubiquitin-specific protease 7), resulting in significantly diminished TRIM27 protein. Loss of WNK1 disrupted WASH ubiquitination and endosomal actin polymerization, which are necessary for endosomal trafficking. Sustained receptor tyrosine kinase (RTK) expression has long been recognized as a key oncogenic signal for the development and growth of human malignancies. Depletion of either WNK1 or TRIM27 significantly increased degradation of the epidermal growth factor receptor (EGFR) following ligand stimulation in breast and lung cancer cells. Like the EGFR, the RTK AXL was also affected similarly by WNK1 depletion but not by inhibition of WNK1 kinase activity. This study uncovers a mechanistic connection between WNK1 and the TRIM27-USP7 axis and extends our fundamental knowledge about the endocytic pathway regulating cell surface receptors.


Assuntos
Actinas , Endossomos , Humanos , Peptidase 7 Específica de Ubiquitina , Fatores de Transcrição , Receptores ErbB , Receptores Proteína Tirosina Quinases , Proteínas de Ligação a DNA , Proteínas Nucleares , Proteína Quinase 1 Deficiente de Lisina WNK
3.
Front Cell Dev Biol ; 10: 935318, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35813203

RESUMO

Metastasis is the major cause of mortality in cancer patients. Analyses of mouse models and patient data have implicated the protein kinase WNK1 as one of a handful of genes uniquely linked to a subset of invasive cancers. WNK1 signaling pathways are widely implicated in the regulation of ion co-transporters and in controlling cell responses to osmotic stress. In this review we will discuss its actions in tumor malignancy in human cancers and present evidence for its function in invasion, migration, angiogenesis and mesenchymal transition.

4.
Am J Physiol Cell Physiol ; 322(6): C1176-C1186, 2022 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-35442829

RESUMO

The with no lysine (K) 1 (WNK1) protein kinase maintains cellular ion homeostasis in many tissues through actions on ion cotransporters and channels. Increased accumulation of WNK1 protein leads to pseudohypoaldosteronism type II (PHAII), a form of familial hypertension. WNK1 can be degraded via its adaptor-dependent recruitment to the Cullin3-RBX1 E3 ligase complex by the ubiquitin-proteasome system. Disruption of this process also leads to disease. To determine if this is the primary mechanism of WNK1 turnover, we examined WNK1 protein stability and degradation by measuring its rate of decay after blockade of translation. Here, we show that WNK1 protein degradation exhibits atypical kinetics in HeLa cells. Consistent with this apparent complexity, we found that multiple degradative pathways can modulate cellular WNK1 protein amount. WNK1 protein is degraded by not only the proteasome but also the lysosome. Non-lysosomal cysteine proteases calpain and caspases also influence WNK1 degradation, as inhibitors of these proteases modestly increased WNK1 protein expression. Importantly, we discovered that the E3 ubiquitin ligase UBR5 interacts with WNK1 and its deficiency results in increased WNK1 protein. Our results further demonstrate that increased WNK1 in UBR5-depleted cells is attributable to reduced lysosomal degradation of WNK1 protein. Taken together, our findings provide insights into the multiplicity of degradative pathways involved in WNK1 turnover and uncover UBR5 as a previously unknown regulator of WNK1 protein stability that leads to lysosomal degradation of WNK1 protein.


Assuntos
Proteínas Serina-Treonina Quinases , Pseudo-Hipoaldosteronismo , Células HeLa , Humanos , Antígenos de Histocompatibilidade Menor/genética , Complexo de Endopeptidases do Proteassoma , Proteínas Serina-Treonina Quinases/genética , Pseudo-Hipoaldosteronismo/metabolismo , Ubiquitina-Proteína Ligases/genética , Ubiquitina-Proteína Ligases/metabolismo , Proteína Quinase 1 Deficiente de Lisina WNK/genética , Proteína Quinase 1 Deficiente de Lisina WNK/metabolismo
5.
Mol Cancer Ther ; 20(10): 1800-1808, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34253593

RESUMO

Metastasis is the major cause of mortality in patients with breast cancer. Many signaling pathways have been linked to cancer invasiveness, but blockade of few protein components has succeeded in reducing metastasis. Thus, identification of proteins contributing to invasion that are manipulable by small molecules may be valuable in inhibiting spread of the disease. The protein kinase with no lysine (K) 1 (WNK1) has been suggested to induce migration of cells representing a range of cancer types. Analyses of mouse models and patient data have implicated WNK1 as one of a handful of genes uniquely linked to invasive breast cancer. Here, we present evidence that inhibition of WNK1 slows breast cancer metastasis. We show that depletion or inhibition of WNK1 reduces migration of several breast cancer cell lines in wound healing assays and decreases invasion in collagen matrices. Furthermore, WNK1 depletion suppresses expression of AXL, a tyrosine kinase implicated in metastasis. Finally, we demonstrate that WNK inhibition in mice attenuates tumor progression and metastatic burden. These data showing reduced migration, invasion, and metastasis upon WNK1 depletion in multiple breast cancer models suggest that WNK1 contributes to the metastatic phenotype, and that WNK1 inhibition may offer a therapeutic avenue for attenuating progression of invasive breast cancers.


Assuntos
Biomarcadores Tumorais/metabolismo , Neoplasias da Mama/patologia , Regulação Neoplásica da Expressão Gênica , Proteína Quinase 1 Deficiente de Lisina WNK/metabolismo , Animais , Apoptose , Biomarcadores Tumorais/genética , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/genética , Neoplasias da Mama/metabolismo , Movimento Celular , Proliferação de Células , Feminino , Humanos , Imidazóis/farmacologia , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Invasividade Neoplásica , Pirrolidinas/farmacologia , Células Tumorais Cultivadas , Proteína Quinase 1 Deficiente de Lisina WNK/antagonistas & inibidores , Proteína Quinase 1 Deficiente de Lisina WNK/genética , Ensaios Antitumorais Modelo de Xenoenxerto
6.
Cell Death Dis ; 12(3): 271, 2021 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-33723235

RESUMO

Cancers, including glioblastoma multiforme (GBM), undergo coordinated reprogramming of metabolic pathways that control glycolysis and oxidative phosphorylation (OXPHOS) to promote tumor growth in diverse tumor microenvironments. Adaptation to limited nutrient availability in the microenvironment is associated with remodeling of mitochondrial morphology and bioenergetic capacity. We recently demonstrated that NF-κB-inducing kinase (NIK) regulates mitochondrial morphology to promote GBM cell invasion. Here, we show that NIK is recruited to the outer membrane of dividing mitochondria with the master fission regulator, Dynamin-related protein1 (DRP1). Moreover, glucose deprivation-mediated metabolic shift to OXPHOS increases fission and mitochondrial localization of both NIK and DRP1. NIK deficiency results in decreased mitochondrial respiration, ATP production, and spare respiratory capacity (SRC), a critical measure of mitochondrial fitness. Although IκB kinase α and ß (IKKα/ß) and NIK are required for OXPHOS in high glucose media, only NIK is required to increase SRC under glucose deprivation. Consistent with an IKK-independent role for NIK in regulating metabolism, we show that NIK phosphorylates DRP1-S616 in vitro and in vivo. Notably, a constitutively active DRP1-S616E mutant rescues oxidative metabolism, invasiveness, and tumorigenic potential in NIK-/- cells without inducing IKK. Thus, we establish that NIK is critical for bioenergetic stress responses to promote GBM cell pathogenesis independently of IKK. Our data suggest that targeting NIK may be used to exploit metabolic vulnerabilities and improve therapeutic strategies for GBM.


Assuntos
Neoplasias Encefálicas/enzimologia , Metabolismo Energético , Glioblastoma/enzimologia , Mitocôndrias/enzimologia , Proteínas Serina-Treonina Quinases/metabolismo , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patologia , Linhagem Celular Tumoral , Dinaminas/genética , Dinaminas/metabolismo , Regulação Neoplásica da Expressão Gênica , Glioblastoma/genética , Glioblastoma/patologia , Humanos , Mitocôndrias/genética , Mitocôndrias/patologia , Dinâmica Mitocondrial , Membranas Mitocondriais/enzimologia , Membranas Mitocondriais/patologia , Fosforilação , Proteínas Serina-Treonina Quinases/genética , Microambiente Tumoral , Quinase Induzida por NF-kappaB
7.
J Cell Biol ; 216(3): 761-778, 2017 03 06.
Artigo em Inglês | MEDLINE | ID: mdl-28193701

RESUMO

Bone homeostasis depends on the functional balance of osteoblasts (OBs) and osteoclasts (OCs). Lrp4 is a transmembrane protein that is mutated in patients with high bone mass. Loss of Lrp4 in OB-lineage cells increases bone mass by elevating bone formation by OBs and reducing bone resorption by OCs. However, it is unclear how Lrp4 deficiency in OBs impairs osteoclastogenesis. Here, we provide evidence that loss of Lrp4 in the OB lineage stabilizes the prorenin receptor (PRR) and increases PRR/V-ATPase-driven ATP release, thereby enhancing the production of the ATP derivative adenosine. Both pharmacological and genetic inhibition of adenosine-2A receptor (A2AR) in culture and Lrp4 mutant mice diminishes the osteoclastogenic deficit and reduces trabecular bone mass. Furthermore, elevated adenosine-A2AR signaling reduces receptor activator of nuclear factor κB (RANK)-mediated osteoclastogenesis. Collectively, these results identify a mechanism by which osteoblastic Lrp4 controls osteoclastogenesis, reveal a cross talk between A2AR and RANK signaling in osteoclastogenesis, and uncover an unrecognized pathophysiological mechanism of high-bone-mass disorders.


Assuntos
Trifosfato de Adenosina/metabolismo , Proteínas Relacionadas a Receptor de LDL/metabolismo , Osteoblastos/metabolismo , Osteogênese/fisiologia , Receptor A2A de Adenosina/metabolismo , Células 3T3 , Animais , Reabsorção Óssea/metabolismo , Reabsorção Óssea/fisiopatologia , Osso e Ossos/metabolismo , Osso e Ossos/fisiologia , Diferenciação Celular/fisiologia , Linhagem Celular , Linhagem da Célula , Células HEK293 , Humanos , Camundongos , Osteoblastos/fisiologia , Osteoclastos/metabolismo , Osteoclastos/fisiologia , Osteoprotegerina/metabolismo , Ligante RANK/metabolismo , Receptor Ativador de Fator Nuclear kappa-B/metabolismo , Transdução de Sinais/fisiologia
8.
Curr Biol ; 26(24): 3288-3302, 2016 12 19.
Artigo em Inglês | MEDLINE | ID: mdl-27889261

RESUMO

Although the role of NF-κB-inducing kinase (NIK) in immunity is well established, its relevance in cancer is just emerging. Here we describe novel functions for NIK in regulating mitochondrial dynamics and motility to promote cell invasion. We show that NIK is localized to mitochondria in cancer cell lines, ex vivo tumor tissue, and mouse embryonic fibroblasts (MEFs). NIK promotes mitochondrial fission, velocity, and directional migration, resulting in subcellular distribution of mitochondria to the periphery of migrating cells. Moreover, NIK is required for recruitment of Drp1 to mitochondria, forms a complex with Drp1, and regulates Drp1 phosphorylation at Ser-616 and dephosphorylation at Ser-637. Consistent with a role for NIK in regulating mitochondrial dynamics, we demonstrate that Drp1 is required for NIK-dependent, cytokine-induced invasion. Importantly, using MEFs, we demonstrate that the established downstream mediators of NIK signaling, IκB kinase α/ß (IKKα/ß) and NF-κB, are not required for NIK to regulate cell invasion, Drp1 mitochondrial localization, or mitochondrial fission. Our results establish a new paradigm for IKK-independent NIK signaling and significantly expand the current dogma that NIK is predominantly cytosolic and exclusively regulates NF-κB activity. Overall, these findings highlight the importance of NIK in tumor pathogenesis and invite new therapeutic strategies that attenuate mitochondrial dysfunction through inhibition of NIK and Drp1.


Assuntos
Fibroblastos/metabolismo , Mitocôndrias/metabolismo , Invasividade Neoplásica , Proteínas Serina-Treonina Quinases/metabolismo , Transporte Proteico/fisiologia , Animais , Linhagem Celular Tumoral , Regulação Enzimológica da Expressão Gênica , Regulação Neoplásica da Expressão Gênica/fisiologia , Humanos , Camundongos , Proteínas Serina-Treonina Quinases/genética , Quinase Induzida por NF-kappaB
9.
Proc Natl Acad Sci U S A ; 112(11): 3487-92, 2015 Mar 17.
Artigo em Inglês | MEDLINE | ID: mdl-25733894

RESUMO

Bone mass is maintained by balanced activity of osteoblasts and osteoclasts. Lrp4 (low-density lipoprotein receptor related protein 4) is a member of the LDL receptor family, whose mutations have been identified in patients with high-bone-mass disorders, such as sclerosteosis and van Buchem diseases. However, it remains unknown whether and how Lrp4 regulates bone-mass homeostasis in vivo. Here we provide evidence that Lrp4-null mutation or specific mutation in osteoblast-lineage cells increased cortical and trabecular bone mass, which was associated with elevated bone formation and impaired bone resorption. This phenotype was not observed in osteoclast-selective Lrp4 knockout mice. Mechanistic studies indicate that loss of Lrp4 function in osteoblast-lineage cells increased serum levels of sclerostin, a key factor for bone-mass homeostasis that interacts with Lrp4, but abolished the inhibition of Wnt/ß-catenin signaling and osteoblastic differentiation by sclerostin. Concomitantly, sclerostin induction of RANKL (receptor activator of nuclear kappa B ligand) was impaired, leading to a lower ratio of RANKL over OPG (osteoprotegerin) (a key factor for osteoclastogenesis). Taken together, these results support the view for Lrp4 as a receptor of sclerostin to inhibit Wnt/ß-catenin signaling and bone formation and identify Lrp4 as a critical player in bone-mass homeostasis.


Assuntos
Reabsorção Óssea/metabolismo , Reabsorção Óssea/patologia , Osteoblastos/metabolismo , Osteoclastos/metabolismo , Osteogênese , Receptores de LDL/metabolismo , Proteínas Adaptadoras de Transdução de Sinal , Aminoácidos/sangue , Animais , Células da Medula Óssea/metabolismo , Células da Medula Óssea/patologia , Reabsorção Óssea/sangue , Diferenciação Celular , Linhagem da Célula , Fêmur/diagnóstico por imagem , Fêmur/patologia , Glicoproteínas/sangue , Glicoproteínas/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular , Proteínas Relacionadas a Receptor de LDL , Camundongos Knockout , Músculos/metabolismo , Especificidade de Órgãos , Osteoblastos/patologia , Osteocalcina/metabolismo , Osteoclastos/patologia , Osteoprotegerina/metabolismo , Ligante RANK/metabolismo , Receptores de LDL/deficiência , Células Estromais/metabolismo , Células Estromais/patologia , Via de Sinalização Wnt , Microtomografia por Raio-X , beta Catenina/metabolismo
10.
Mol Cancer ; 14: 9, 2015 Jan 27.
Artigo em Inglês | MEDLINE | ID: mdl-25622756

RESUMO

BACKGROUND: High-grade gliomas are one of the most invasive and therapy-resistant cancers. We have recently shown that noncanonical NF-κB/RelB signaling is a potent driver of tumorigenesis and invasion in the aggressive, mesenchymal subtype of glioma. However, the relevant signals that induce activation of noncanonical NF-κB signaling in glioma and its function relative to the canonical NF-κB pathway remain elusive. METHODS: The ability of tumor necrosis factor (TNF)-like weak inducer of apoptosis (TWEAK) to regulate NF-κB signaling and promote tumor progression was investigated in both established and primary high-grade glioma tumor lines using a three-dimensional (3-D) collagen invasion assay. The roles of specific NF-κB proteins in regulating glioma cell invasion and expression of Matrix Metalloproteinase 9 (MMP9) in response to TWEAK were evaluated using shRNA-mediated loss-of-function studies. The ability of NF-κB-inducing kinase (NIK) to promote glioma growth in vivo was investigated using an orthotopic xenograft mouse model. RESULTS: In glioma cells that display elevated noncanonical NF-κB signaling, loss of RelB attenuates invasion without affecting RelA expression or phosphorylation and RelB is sufficient to promote invasion in the absence of RelA. The cytokine TWEAK preferentially activates the noncanonical NF-κB pathway through induction of p100 processing to p52 and nuclear accumulation of both RelB and p52 without activating the canonical NF-κB pathway. Moreover, TWEAK, but not TNFα, significantly increases NIK mRNA levels. TWEAK also promotes noncanonical NFκB-dependent MMP9 expression and glioma cell invasion. Finally, expression of NIK is sufficient to increase gliomagenesis in vivo. CONCLUSIONS: Our data establish a key role for NIK and noncanonical NF-κB in mediating TWEAK-induced, MMP-dependent glioma cell invasion. The findings also demonstrate that TWEAK induces noncanonical NF-κB signaling and signal-specific regulation of NIK mRNA expression. Together, these studies reveal the important role of noncanonical NF-κB signaling in regulating glioma invasiveness and highlight the therapeutic potential of targeting activation of NIK in this deadly disease.


Assuntos
Glioma/genética , Glioma/patologia , NF-kappa B/genética , Invasividade Neoplásica/genética , Proteínas Serina-Treonina Quinases/genética , Transdução de Sinais/genética , Fatores de Necrose Tumoral/genética , Animais , Apoptose/genética , Linhagem Celular , Linhagem Celular Tumoral , Citocina TWEAK , Células HEK293 , Humanos , Metaloproteinase 9 da Matriz/genética , Camundongos , Camundongos Nus , Invasividade Neoplásica/patologia , Fosforilação/genética , RNA Mensageiro/genética , RNA Interferente Pequeno/genética , Quinase Induzida por NF-kappaB
11.
J Bone Miner Res ; 28(10): 2122-35, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23649480

RESUMO

Reduced bone mineral density and hip fracture are frequently observed in patients with Alzheimer's disease (AD). However, mechanisms underlying their association remain poorly understood. Amyloid precursor protein (APP) is a transmembrane protein that is ubiquitously expressed in bone marrow stromal cells (BMSCs), osteoblasts (OBs), macrophages (BMMs), and osteoclasts (OCs). Mutations in the APP gene identified in early-onset AD patients are believed to cause AD. But little is known about APP's role in bone remodeling. Here, we present evidence for Swedish mutant APP (APPswe) in suppression of OB differentiation and function in culture and in mouse. APP expression in BMSCs increases during aging. Ubiquitous expression of APPswe in young adult Tg2576 transgenic mice (under the control of a prion promoter) recaptured skeletal "aging-like" deficits, including decreased OB genesis and bone formation, increased adipogenesis and bone marrow fat, and enhanced OC genesis and bone resorption. Remarkably, selective expression of APPswe in mature OB-lineage cells in TgAPPswe-Ocn mice (under the control of osteocalcin [Ocn] promoter-driven Cre) also decreased OB genesis and increased OC formation, resulting in a trabecular bone loss. These results thus suggest a cell-autonomous role for APPswe in suppressing OB formation and function, but a nonautonomous effect on OC genesis. Notably, increased adipogenesis and elevated bone marrow fat were detected in young adult Tg2576 mice, but not in TgAPPswe-Ocn mice, implying that APPswe in BMSCs and/or multicell types in bone marrow promotes bone marrow adipogenesis. Intriguingly, the skeletal aging-like deficits in young adult Tg2576 mice were prevented by treatment with N-acetyl-L-cysteine (NAC), an antioxidant, suggesting that reactive oxygen species (ROS) may underlie APPswe-induced osteoporotic deficits. Taken together, these results demonstrate a role for APPswe in suppressing OB differentiation and bone formation, implicate APPswe as a detrimental factor for AD-associated osteoporotic deficit, and reveal a potential clinical value of NAC in the treatment of osteoporotic deficits. © 2013 American Society for Bone and Mineral Research.


Assuntos
Acetilcisteína/uso terapêutico , Peptídeos beta-Amiloides/metabolismo , Diferenciação Celular , Mutação/genética , Osteoblastos/patologia , Osteoporose/tratamento farmacológico , Osteoporose/patologia , Acetilcisteína/farmacologia , Fosfatase Ácida/metabolismo , Adipogenia/efeitos dos fármacos , Envelhecimento/patologia , Animais , Animais Recém-Nascidos , Reabsorção Óssea/complicações , Reabsorção Óssea/tratamento farmacológico , Reabsorção Óssea/metabolismo , Reabsorção Óssea/patologia , Osso e Ossos/efeitos dos fármacos , Osso e Ossos/metabolismo , Osso e Ossos/patologia , Diferenciação Celular/efeitos dos fármacos , Linhagem da Célula/efeitos dos fármacos , Células Cultivadas , Cricetinae , Humanos , Isoenzimas/metabolismo , Células-Tronco Mesenquimais/efeitos dos fármacos , Células-Tronco Mesenquimais/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Tamanho do Órgão/efeitos dos fármacos , Osteoblastos/efeitos dos fármacos , Osteoblastos/metabolismo , Osteogênese/efeitos dos fármacos , Osteoporose/complicações , Osteoporose/metabolismo , Fosfatase Ácida Resistente a Tartarato
12.
J Cell Biol ; 200(6): 821-37, 2013 Mar 18.
Artigo em Inglês | MEDLINE | ID: mdl-23509071

RESUMO

Receptor activator of NF-κB (RANK) plays a critical role in osteoclastogenesis, an essential process for the initiation of bone remodeling to maintain healthy bone mass and structure. Although the signaling and function of RANK have been investigated extensively, much less is known about the negative regulatory mechanisms of its signaling. We demonstrate in this paper that RANK trafficking, signaling, and function are regulated by VPS35, a major component of the retromer essential for selective endosome to Golgi retrieval of membrane proteins. VPS35 loss of function altered RANK ligand (RANKL)-induced RANK distribution, enhanced RANKL sensitivity, sustained RANKL signaling, and increased hyperresorptive osteoclast (OC) formation. Hemizygous deletion of the Vps35 gene in mice promoted hyperresorptive osteoclastogenesis, decreased bone formation, and caused a subsequent osteoporotic deficit, including decreased trabecular bone volumes and reduced trabecular thickness and density in long bones. These results indicate that VPS35 critically deregulates RANK signaling, thus restraining increased formation of hyperresorptive OCs and preventing osteoporotic deficits.


Assuntos
Osteoclastos/metabolismo , Osteoporose/metabolismo , Ligante RANK/metabolismo , Transdução de Sinais , Proteínas de Transporte Vesicular/metabolismo , Animais , Osso e Ossos/metabolismo , Osso e Ossos/patologia , Complexo de Golgi/genética , Complexo de Golgi/metabolismo , Camundongos , Camundongos Mutantes , Tamanho do Órgão , Osteoclastos/patologia , Osteoporose/genética , Osteoporose/patologia , Transporte Proteico/genética , Ligante RANK/genética , Receptor Ativador de Fator Nuclear kappa-B/genética , Receptor Ativador de Fator Nuclear kappa-B/metabolismo , Proteínas de Transporte Vesicular/genética
13.
J Cell Biol ; 195(5): 765-79, 2011 Nov 28.
Artigo em Inglês | MEDLINE | ID: mdl-22105352

RESUMO

VPS35, a major component of the retromer complex, is important for endosome-to-Golgi retrieval of membrane proteins. Although implicated in Alzheimer's disease (AD), how VPS35 regulates AD-associated pathology is unknown. In this paper, we show that hemizygous deletion of Vps35 in the Tg2576 mouse model of AD led to earlier-onset AD-like phenotypes, including cognitive memory deficits, defective long-term potentiation, and impaired postsynaptic glutamatergic neurotransmission in young adult age. These deficits correlated well with an increase of ß-amyloid peptide (Aß) level in the mutant hippocampus. We further demonstrate that VPS35 is predominantly expressed in pyramidal neurons of young adult hippocampus and interacts with BACE1, a protease responsible for Aß production. Loss of VPS35 function in the mouse hippocampus increased BACE1 activity. Suppression of VPS35 expression in culture decreased BACE1 trans-Golgi localization but enriched it in endosomes. These results demonstrate an essential role for VPS35 in suppression of AD neuropathology and in inhibition of BACE1 activation and Aß production by promoting BACE1 endosome-to-Golgi retrieval.


Assuntos
Doença de Alzheimer/genética , Haploinsuficiência , Proteínas de Transporte Vesicular/genética , Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Secretases da Proteína Precursora do Amiloide/metabolismo , Peptídeos beta-Amiloides/metabolismo , Animais , Ácido Aspártico Endopeptidases/metabolismo , Córtex Cerebral/metabolismo , Endossomos/metabolismo , Células HEK293 , Hipocampo/metabolismo , Humanos , Camundongos , Proteínas de Transporte Vesicular/metabolismo , Proteínas de Transporte Vesicular/fisiologia , Rede trans-Golgi/metabolismo
14.
J Bone Miner Res ; 26(5): 1084-98, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21542009

RESUMO

Alzheimer's disease (AD), one of the most dreaded neurodegenerative disorders, is characterized by cortical and cerebrovascular amyloid ß peptide (Aß) deposits, neurofibrillary tangles, chronic inflammation, and neuronal loss. Increased bone fracture rates and reduced bone density are commonly observed in patients with AD, suggesting one or more common denominators between both disorders. However, very few studies are available that have addressed this issue. Here, we present evidence for a function of amyloid precursor protein (APP) and Aß in regulating osteoclast (OC) differentiation in vitro and in vivo. Tg2576 mice, which express the Swedish mutation of APP (APPswe) under the control of a prion promoter, exhibit biphasic effects on OC activation, with an increase of OCs in younger mice (< 4 months old), but a decrease in older Tg2576 mice (> 4 months old). The increase of OCs in young Tg2576 mice appears to be mediated by Aß oligomers and receptor for advanced glycation end products (RAGE) expression in bone marrow macrophages (BMMs). However, the decrease of OC formation and activity in older Tg2576 mice may be due to the increase of soluble rage (sRAGE) in aged Tg2576 mice, an inhibitor of RANKL-induced osteoclastogenesis. These results suggest an unexpected function of APPswe/Aß, reveal a mechanism underlying altered bone remodeling in AD patients, and implicate APP/Aß and RAGE as common denominators for both AD and osteoporosis.


Assuntos
Envelhecimento/metabolismo , Peptídeos beta-Amiloides/farmacologia , Mutação/genética , Osteoblastos/citologia , Osteoblastos/metabolismo , Receptores Imunológicos/metabolismo , Envelhecimento/efeitos dos fármacos , Peptídeos beta-Amiloides/química , Animais , Células da Medula Óssea/citologia , Células da Medula Óssea/efeitos dos fármacos , Células da Medula Óssea/metabolismo , Osso e Ossos/anatomia & histologia , Osso e Ossos/efeitos dos fármacos , Osso e Ossos/metabolismo , Humanos , Injeções Intraperitoneais , Camundongos , Camundongos Transgênicos , Modelos Biológicos , Tamanho do Órgão , Especificidade de Órgãos/efeitos dos fármacos , Osteoblastos/efeitos dos fármacos , Osteoclastos/citologia , Osteoclastos/efeitos dos fármacos , Osteoclastos/metabolismo , Osteogênese/efeitos dos fármacos , Estrutura Quaternária de Proteína , Ligante RANK/farmacologia , Receptor para Produtos Finais de Glicação Avançada , Receptores Imunológicos/administração & dosagem , Solubilidade/efeitos dos fármacos
15.
Blood ; 115(15): 3136-45, 2010 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-20065295

RESUMO

Neogenin, a deleted in colorectal cancer (DCC) family member, has been identified as a receptor for the neuronal axon guidance cues netrins and repulsive guidance molecules repulsive guidance molecules (RGM). RGMc, also called hemojuvelin (HJV), is essential for iron homeostasis. Here we provide evidence that neogenin plays a critical role in iron homeostasis by regulation of HJV secretion and bone morphogenetic protein (BMP) signaling. Livers of neogenin mutant mice exhibit iron overload, low levels of hepcidin, and reduced BMP signaling. Mutant hepatocytes in vitro show impaired BMP2 induction of Smad1/5/8 phosphorylation and hepcidin expression. Neogenin is expressed in liver cells in a reciprocal pattern to that of hepcidin, suggesting that neogenin functions in a cell nonautonomous manner. Further studies demonstrate that neogenin may stabilize HJV, a glycosylphosphatidylinositol-anchored protein that interacts with neogenin and suppresses its secretion. Taken together, our results lead the hypothesis that neogenin regulates iron homeostasis via inhibiting secretion of HJV, an inhibitor of BMP signaling, to enhance BMP signaling and hepcidin expression. These results reveal a novel mechanism underlying neogenin regulation of HJV-BMP signaling.


Assuntos
Peptídeos Catiônicos Antimicrobianos/metabolismo , Proteína Morfogenética Óssea 2/farmacologia , Homeostase/efeitos dos fármacos , Ferro/metabolismo , Proteínas de Membrana/metabolismo , Animais , Peptídeos Catiônicos Antimicrobianos/genética , Proteínas de Transporte de Cátions/metabolismo , Espaço Extracelular/efeitos dos fármacos , Espaço Extracelular/metabolismo , Proteínas Ligadas por GPI , Regulação da Expressão Gênica/efeitos dos fármacos , Proteína da Hemocromatose , Células Hep G2 , Hepatócitos/efeitos dos fármacos , Hepatócitos/metabolismo , Hepatócitos/patologia , Hepcidinas , Humanos , Sobrecarga de Ferro/genética , Sobrecarga de Ferro/patologia , Fígado/efeitos dos fármacos , Fígado/metabolismo , Fígado/patologia , Proteínas de Membrana/deficiência , Proteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Fosforilação/efeitos dos fármacos , Transporte Proteico/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Proteínas Smad/metabolismo
16.
Exp Mol Med ; 41(12): 935-45, 2009 Dec 31.
Artigo em Inglês | MEDLINE | ID: mdl-19745600

RESUMO

Glycosphingolipids including gangliosides play important regulatory roles in cell proliferation and differentiation. UDP-glucose:ceramide glucosyltransferase (Ugcg) catalyze the initial step in glycosphingolipids biosynthesis pathway. In this study, Ugcg expression was reduced to approximately 80% by short hairpin RNAs (shRNAs) to evaluate the roles of glycosphingolipids in proliferation and neural differentiation of mouse embryonic stem cells (mESCs). HPTLC/immunofluorescence analyses of shRNA- transfected mESCs revealed that treatment with Ugcg-shRNA decreased expression of major gangliosides, GM3 and GD3. Furthermore, MTT and Western blot/immunofluorescence analyses demonstrated that inhibition of the Ugcg expression in mESCs resulted in decrease of cell proliferation (P<0.05) and decrease of activation of the ERK1/2 (P<0.05), respectively. To further investigate the role of glycosphingolipids in neural differentiation, the embryoid bodies formed from Ugcg-shRNA transfected mESCs were differentiated into neural cells by treatment with retinoic acid. We found that inhibition of Ugcg expression did not affect embryoid body (EB) differentiation, as judged by morphological comparison and expression of early neural precursor cell marker, nestin, in differentiated EBs. However, RT-PCR/immunofluorescence analyses showed that expression of microtubule-associated protein 2 (MAP-2) for neurons and glial fibrillary acidic protein (GFAP) for glial cells was decreased in neural cells differentiated from the shRNA-transfected mESCs. These results suggest that glycosphingolipids are involved in the proliferation of mESCs through ERK1/2 activation, and that glycosphingolipids play roles in differentiation of neural precursor cells derived from mESCs.


Assuntos
Proliferação de Células , Células-Tronco Embrionárias/citologia , Glicoesfingolipídeos/metabolismo , Neurogênese , Neurônios/citologia , Animais , Células Cultivadas , Regulação para Baixo , Células-Tronco Embrionárias/metabolismo , Glucosiltransferases/genética , Glucosiltransferases/metabolismo , Glicoesfingolipídeos/genética , Camundongos , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Neurônios/metabolismo , RNA Mensageiro/genética
17.
Biochem Biophys Res Commun ; 371(4): 866-71, 2008 Jul 11.
Artigo em Inglês | MEDLINE | ID: mdl-18471991

RESUMO

Gangliosides are sialic acid-conjugated glycosphingolipids that are believed to regulate cell differentiation as well as the signals of several signal molecules, including epidermal growth factor receptors (EGFR). These compounds are localized in a glycosphingolipid-enriched microdomain on the cell surface and regulated by the glycosphingolipid composition. However, the role that gangliosides play in osteoblastogenesis is not yet clearly understood, therefore, in this study, the relationship between gangliosides and EGFR activation was investigated during osteoblast differentiation in human mesenchymal stem cells (hMSCs). The results of high-performance thin-layer chromatography (HPTLC) showed that ganglioside GM3 expression was decreased, whereas ganglioside GD1a expression was increased during the differentiation of hMSCs into osteoblasts. In addition, an increase in the activation of alkaline phosphatase (ALP) was observed in response to treatment with EGF (5 ng/ml) and GD1a (1 microM) (p<0.05). The activation of ALP was significantly elevated in response to treatment of ganglioside GD1a with EGF when compared to control cells (p<0.01). However, treatment with GM3 (1muM) resulted in decreased ALP activation (p<0.01), and treatment of hMSCs with a chemical inhibitor of EGFR, AG1478, removed the differential effect of the two gangliosides. Moreover, incubation of the differentiating cells with GD1a enhanced the phosphorylation of EGFR, whereas treatment with GM3 reduced the EGFR phosphorylation. However, AG1478 treatment inhibited the effect of ganglioside GD1a elicitation on EGFR phosphorylation. Taken together, these results indicate that GD1a promotes osteoblast differentiation through the enhancement of EGFR phosphorylation, but that GM3 inhibits osteoblast differentiation through reduced EGFR phosphorylation, suggesting that GM3 and GD1a are essential molecules for regulating osteoblast differentiation in hMSCs.


Assuntos
Diferenciação Celular , Receptores ErbB/metabolismo , Gangliosídeo G(M3)/fisiologia , Gangliosídeos/fisiologia , Células-Tronco Mesenquimais/citologia , Osteoblastos/citologia , Fosfatase Alcalina/metabolismo , Diferenciação Celular/efeitos dos fármacos , Fator de Crescimento Epidérmico/farmacologia , Receptores ErbB/agonistas , Gangliosídeo G(M3)/farmacologia , Gangliosídeos/farmacologia , Humanos , Células-Tronco Mesenquimais/efeitos dos fármacos , Células-Tronco Mesenquimais/metabolismo , Osteoblastos/metabolismo , Fosforilação , Inibidores de Proteínas Quinases/farmacologia , Quinazolinas , Tirfostinas/farmacologia
18.
Arch Pharm Res ; 31(1): 88-95, 2008 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18277613

RESUMO

Gangliosides are a family of sialic acid-containing glycosphingolipids that are abundant in neurons and have a variety of functions in developing and mature tissues. We examined the expression of ganglioside GT1b in the embryonic preimplantation stage after freezing and thawing processes to determine the regulatory roles of ganglioside GT1b in early embryonic development. ICR mouse embryos at the two-cell stage obtained by flushing the oviducts were frozen by two cryopreservation procedures, slow freezing using a programmable freezer or vitrification by direct plunging into liquid nitrogen. Slow freezing was conducted with equilibration in 1.5 M 1,2-propanediol or 5% equilibration glycerol. Vitrification was applied with a 10-15 min equilibration in 7.5% ethylene glycol (EG), 7.5% dimethylsulfoxide (DMSO), and 30 sec in a solution of 15% EG, 15% DMSO and 0.5 M sucrose. Immediately after thawing, the survival rate of the embryos was assessed by their morphology and ability to develop to blastocysts in culture. The survival rate of vitrified and thawed embryos (92%) was significantly higher than that of slow frozen and thawed embryos (76%) (P<0.05). A tendency of higher blastocyst rate was found in the vitrified and thawed embryos compared to that of the slow frozen and thawed embryos. Confocal immunofluorescence staining confirmed that surviving embryos expressed ganglioside GT1b, with the strongest expression at the compacted eight-cell or later stage embryos. Ganglioside GT1b was not observed in the TUNEL-positive, apoptotic embryos, suggesting that cryopreservation had induced DNA breaks in them. These results suggest that ganglioside GT1b may play an important role in embryo survival or development.


Assuntos
Desenvolvimento Embrionário/fisiologia , Gangliosídeos/biossíntese , Animais , Apoptose/efeitos dos fármacos , Blastocisto/metabolismo , Sobrevivência Celular/efeitos dos fármacos , Criopreservação , Feminino , Imunofluorescência , Marcação In Situ das Extremidades Cortadas , Masculino , Camundongos , Camundongos Endogâmicos ICR , Mórula/metabolismo , Gravidez , Zona Pelúcida/efeitos dos fármacos , Zona Pelúcida/ultraestrutura
19.
Biochem Biophys Res Commun ; 362(2): 313-8, 2007 Oct 19.
Artigo em Inglês | MEDLINE | ID: mdl-17707770

RESUMO

Gangliosides are implicated in neuronal development processes. The regulation of ganglioside levels is closely related to the induction of neuronal cell differentiation. In this study, the relationship between ganglioside expression and neuronal cell development was investigated using an in vitro model of neural differentiation from mouse embryonic stem (mES) cells. Daunorubicin (DNR) was applied to induce the expression of gangliosides in embryoid body (EB) (4+). We observed an increase in expression of gangliosides in all stages of EBs by treatment of DNR (2microM). High-performance thin-layer chromatography (HPTLC) showed that gangliosides GD3, GD1a, GT1b, and GQ1b increased in DNR-treated 7-day-old EB (4+) [EB (4+):7]. DNR treatment significantly increased the expression of gangliosides, especially GT1b and GQ1b in comparison to control cells. Interestingly, GQ1b co-localized with microtubule-associated protein 2 (MAP-2) expressing cells in DNR-treated EB (4+):7. The co-localization of GQ1b and MAP-2 was found in neurite-bearing cells in DNR-treated 15-day-old EB (4+) [EB (4+):15], whereas no significant expression of GQ1b and less neurite formation were observed in untreated control. Also, the expression of synaptophysin and NF200, both neuronal markers associated with neruites, was increased by DNR treatment. These results demonstrate that DNR increases expression of gangliosides, especially GQ1b, in differentiating neuronal cells. Further, neurite-bearing neuronal cell differentiation can be facilitated by DNR, possibly through the induction of gangliosides. Thus, the present data suggest that DNR is beneficial for facilitating neuronal differentiation from ES cells and among the gangliosides analyzed in the present study, GQ1b is mainly involved in neurite formation.


Assuntos
Diferenciação Celular/efeitos dos fármacos , Daunorrubicina/farmacologia , Células-Tronco Embrionárias/efeitos dos fármacos , Gangliosídeos/biossíntese , Neurônios/efeitos dos fármacos , Animais , Antibióticos Antineoplásicos/farmacologia , Linhagem Celular , Proliferação de Células/efeitos dos fármacos , Cromatografia Líquida de Alta Pressão , Cromatografia em Camada Fina , Relação Dose-Resposta a Droga , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/metabolismo , Imuno-Histoquímica , Camundongos , Neurônios/citologia , Neurônios/metabolismo , Fatores de Tempo
20.
Arch Pharm Res ; 29(8): 666-76, 2006 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16964762

RESUMO

Gangliosides are widely distributed in mammalian cells and play important roles in various functions such as cell differentiation and growth control. In addition, diabetes and obesity cause abnormal development of reproductive processes in a variety of species. However, the mechanisms underlying these effects, and how they are related, are not fully understood. This study examined whether the differential expression of gangliosides is implicated in the abnormal follicular development and uterine architecture of streptozotocin (STZ)-induced and db/db diabetic mice. Based upon the mobility on high-performance thin-layer chromatography, mouse ovary consisted of at least five different ganglioside components, mainly gangliosides GM3, GM1, GD1a and GT1b, and diabetic ovary exhibited a significant reduction in ganglioside expression with apparent changes in the major gangliosides. A prominent immunofluorescence microscopy showed a dramatic loss of ganglioside GD1a expression in the primary, secondary and Graafian follicles of STZ-induced and db/db diabetic mice. A significant decrease in ganglioside GD3 expression was also observed in the ovary of db/db mice. In the uterus of STZ-induced diabetic mice, expression of gangliosides GD1a and GT1b was obviously reduced, but gangliosides GM1, GM2 and GD3 expression was increased. In contrast, the uterus of db/db mice showed a significant increase in gangliosides GM1, GD1a and GD3 expression. Taken together, a complex pattern of ganglioside expression was seen in the ovary and uterus of normoglycemic ICR and db/+ mice, and the correspoding tissues in diabetic mice are characterized by appreciable changes of the major ganglioside expression. These results suggest that alterations in ganglioside expression caused by diabetes mellitus may be implicated in abnormal ovarian development and uterine structure.


Assuntos
Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Tipo 1/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Gangliosídeos/biossíntese , Ovário/metabolismo , Útero/metabolismo , Animais , Cromatografia em Camada Fina , Diabetes Mellitus Experimental/induzido quimicamente , Diabetes Mellitus Tipo 1/induzido quimicamente , Diabetes Mellitus Tipo 2/genética , Feminino , Camundongos , Camundongos Endogâmicos ICR , Camundongos Mutantes , Microscopia de Fluorescência , Folículo Ovariano/metabolismo , Especificidade da Espécie , Estreptozocina
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...