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











Base de dados
Intervalo de ano de publicação
1.
Semin Cell Dev Biol ; 133: 42-52, 2023 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-35256275

RESUMO

Membrane trafficking is a core cellular process that supports diversification of cell shapes and behaviors relevant to morphogenesis during development and in adult organisms. However, how precisely trafficking components regulate specific differentiation programs is incompletely understood. Snap29 is a multifaceted Soluble N-ethylmaleimide-sensitive factor Attachment protein Receptor, involved in a wide range of trafficking and non-trafficking processes in most cells. A body of knowledge, accrued over more than two decades since its discovery, reveals that Snap29 is essential for establishing and maintaining the operation of a number of cellular events that support cell polarity and signaling. In this review, we first summarize established functions of Snap29 and then we focus on novel ones in the context of autophagy, Golgi trafficking and vesicle fusion at the plasma membrane, as well as on non-trafficking activities of Snap29. We further describe emerging evidence regarding the compartmentalisation and regulation of Snap29. Finally, we explore how the loss of distinct functions of human Snap29 may lead to the clinical manifestations of congenital disorders such as CEDNIK syndrome and how altered SNAP29 activity may contribute to the pathogenesis of cancer, viral infection and neurodegenerative diseases.


Assuntos
Ceratodermia Palmar e Plantar , Síndromes Neurocutâneas , Humanos , Proteínas Qc-SNARE/genética , Proteínas Qc-SNARE/metabolismo , Proteínas Qb-SNARE/genética , Proteínas Qb-SNARE/metabolismo , Ceratodermia Palmar e Plantar/metabolismo , Ceratodermia Palmar e Plantar/patologia , Síndromes Neurocutâneas/metabolismo , Síndromes Neurocutâneas/patologia , Morfogênese
2.
Cell Death Dis ; 13(11): 1003, 2022 11 27.
Artigo em Inglês | MEDLINE | ID: mdl-36435842

RESUMO

The oncoprotein GOLPH3 (Golgi phosphoprotein 3) is an evolutionarily conserved phosphatidylinositol 4-phosphate effector, mainly localized to the Golgi apparatus, where it supports organelle architecture and vesicular trafficking. Overexpression of human GOLPH3 correlates with poor prognosis in several cancer types and is associated with enhanced signaling downstream of mTOR (mechanistic target of rapamycin). However, the molecular link between GOLPH3 and mTOR remains elusive. Studies in Drosophila melanogaster have shown that Translationally controlled tumor protein (Tctp) and 14-3-3 proteins are required for organ growth by supporting the function of the small GTPase Ras homolog enriched in the brain (Rheb) during mTORC1 (mTOR complex 1) signaling. Here we demonstrate that Drosophila GOLPH3 (dGOLPH3) physically interacts with Tctp and 14-3-3ζ. RNAi-mediated knockdown of dGOLPH3 reduces wing and eye size and enhances the phenotypes of Tctp RNAi. This phenotype is partially rescued by overexpression of Tctp, 14-3-3ζ, or Rheb. We also show that the Golgi localization of Rheb in Drosophila cells depends on dGOLPH3. Consistent with dGOLPH3 involvement in Rheb-mediated mTORC1 activation, depletion of dGOLPH3 also reduces levels of phosphorylated ribosomal S6 kinase, a downstream target of mTORC1. Finally, the autophagy flux and the expression of autophagic transcription factors of the TFEB family, which anti correlates with mTOR signaling, are compromised upon reduction of dGOLPH3. Overall, our data provide the first in vivo demonstration that GOLPH3 regulates organ growth by directly associating with mTOR signaling proteins.


Assuntos
Drosophila , Neuropeptídeos , Animais , Humanos , Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Proteína Enriquecida em Homólogo de Ras do Encéfalo/metabolismo , Proteínas 14-3-3/metabolismo , Neuropeptídeos/genética , Serina-Treonina Quinases TOR/genética , Serina-Treonina Quinases TOR/metabolismo , Alvo Mecanístico do Complexo 1 de Rapamicina/metabolismo , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo
3.
Neuropathol Appl Neurobiol ; 48(5): e12818, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-35501124

RESUMO

AIM: Mutations in the valosin-containing protein (VCP) gene cause various lethal proteinopathies that mainly include inclusion body myopathy with Paget's disease of bone and frontotemporal dementia (IBMPFD) and amyotrophic lateral sclerosis (ALS). Different pathological mechanisms have been proposed. Here, we define the impact of VCP mutants on lysosomes and how cellular homeostasis is restored by inducing autophagy in the presence of lysosomal damage. METHODS: By electron microscopy, we studied lysosomal morphology in VCP animal and motoneuronal models. With the use of western blotting, real-time quantitative polymerase chain reaction (RT-qPCR), immunofluorescence and filter trap assay, we evaluated the effect of selected VCP mutants in neuronal cells on lysosome size and activity, lysosomal membrane permeabilization and their impact on autophagy. RESULTS: We found that VCP mutants induce the formation of aberrant multilamellar organelles in VCP animal and cell models similar to those found in patients with VCP mutations or with lysosomal storage disorders. In neuronal cells, we found altered lysosomal activity characterised by membrane permeabilization with galectin-3 redistribution and activation of PPP3CB. This selectively activated the autophagy/lysosomal transcriptional regulator TFE3, but not TFEB, and enhanced both SQSTM1/p62 and lipidated MAP1LC3B levels inducing autophagy. Moreover, we found that wild type VCP, but not the mutants, counteracted lysosomal damage induced either by trehalose or by a mutant form of SOD1 (G93A), also blocking the formation of its insoluble intracellular aggregates. Thus, chronic activation of autophagy might fuel the formation of multilamellar bodies. CONCLUSION: Together, our findings provide insights into the pathogenesis of VCP-related diseases, by proposing a novel mechanism of multilamellar body formation induced by VCP mutants that involves lysosomal damage and induction of lysophagy.


Assuntos
Adenosina Trifosfatases , Proteínas de Ciclo Celular , Adenosina Trifosfatases/genética , Adenosina Trifosfatases/metabolismo , Animais , Autofagia/genética , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Lisossomos/metabolismo , Neurônios Motores/metabolismo , Ativação Transcricional , Proteína com Valosina/genética , Proteína com Valosina/metabolismo
4.
Autophagy ; 17(12): 4442-4452, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-33978540

RESUMO

Glioblastoma (GBM), a very aggressive and incurable tumor, often results from constitutive activation of EGFR (epidermal growth factor receptor) and of phosphoinositide 3-kinase (PI3K). To understand the role of autophagy in the pathogenesis of glial tumors in vivo, we used an established Drosophila melanogaster model of glioma based on overexpression in larval glial cells of an active human EGFR and of the PI3K homolog Pi3K92E/Dp110. Interestingly, the resulting hyperplastic glia express high levels of key components of the lysosomal-autophagic compartment, including vacuolar-type H+-ATPase (V-ATPase) subunits and ref(2)P (refractory to Sigma P), the Drosophila homolog of SQSTM1/p62. However, cellular clearance of autophagic cargoes appears inhibited upstream of autophagosome formation. Remarkably, downregulation of subunits of V-ATPase, of Pdk1, or of the Tor (Target of rapamycin) complex 1 (TORC1) component raptor prevents overgrowth and normalize ref(2)P levels. In addition, downregulation of the V-ATPase subunit VhaPPA1-1 reduces Akt and Tor-dependent signaling and restores clearance. Consistent with evidence in flies, neurospheres from patients with high V-ATPase subunit expression show inhibition of autophagy. Altogether, our data suggest that autophagy is repressed during glial tumorigenesis and that V-ATPase and MTORC1 components acting at lysosomes could represent therapeutic targets against GBM.


Assuntos
Neoplasias , ATPases Vacuolares Próton-Translocadoras , Animais , Autofagia/genética , Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Humanos , Lisossomos/metabolismo , Neoplasias/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , ATPases Vacuolares Próton-Translocadoras/metabolismo
5.
Sci Rep ; 9(1): 1211, 2019 02 04.
Artigo em Inglês | MEDLINE | ID: mdl-30718891

RESUMO

Homozygous mutations in SNAP29, encoding a SNARE protein mainly involved in membrane fusion, cause CEDNIK (Cerebral Dysgenesis, Neuropathy, Ichthyosis and Keratoderma), a rare congenital neurocutaneous syndrome associated with short life expectancy, whose pathogenesis is unclear. Here, we report the analysis of the first genetic model of CEDNIK in zebrafish. Strikingly, homozygous snap29 mutant larvae display CEDNIK-like features, such as microcephaly and skin defects. Consistent with Snap29 role in membrane fusion during autophagy, we observe accumulation of the autophagy markers p62 and LC3, and formation of aberrant multilamellar organelles and mitochondria. Importantly, we find high levels of apoptotic cell death during early development that might play a yet uncharacterized role in CEDNIK pathogenesis. Mutant larvae also display mouth opening problems, feeding impairment and swimming difficulties. These alterations correlate with defective trigeminal nerve formation and excess axonal branching. Since the paralog Snap25 is known to promote axonal branching, Snap29 might act in opposition with, or modulate Snap25 activity during neurodevelopment. Our vertebrate genetic model of CEDNIK extends the description in vivo of the multisystem defects due to loss of Snap29 and could provide the base to test compounds that might ameliorate traits of the disease.


Assuntos
Ceratodermia Palmar e Plantar/metabolismo , Síndromes Neurocutâneas/metabolismo , Proteínas SNARE/metabolismo , Proteínas de Peixe-Zebra/metabolismo , Animais , Autofagia , Humanos , Ceratodermia Palmar e Plantar/genética , Ceratodermia Palmar e Plantar/fisiopatologia , Fusão de Membrana , Modelos Genéticos , Mutação , Malformações do Sistema Nervoso/metabolismo , Síndromes Neurocutâneas/genética , Síndromes Neurocutâneas/fisiopatologia , Fenótipo , Ligação Proteica , Proteínas Qb-SNARE/metabolismo , Proteínas Qc-SNARE/metabolismo , Proteínas SNARE/fisiologia , Proteína 25 Associada a Sinaptossoma/metabolismo , Proteína 25 Associada a Sinaptossoma/fisiologia , Peixe-Zebra/metabolismo , Proteínas de Peixe-Zebra/fisiologia
6.
EBioMedicine ; 41: 225-235, 2019 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-30737083

RESUMO

BACKGROUND: The V-ATPase proton pump controls acidification of intra and extra-cellular milieu in both physiological and pathological conditions. We previously showed that some V-ATPase subunits are enriched in glioma stem cells and in patients with poor survival. In this study, we investigated how expression of a GBM-like V-ATPase pump influences the non-neoplastic brain microenvironment. METHODS: Large oncosome (LO) vesicles were isolated from primary glioblastoma (GBM) neurospheres, or from patient sera, and co-cultured with primary neoplastic or non-neoplastic brain cells. LO transcript and protein contents were analyzed by qPCR, immunoblotting and immunogold staining. Activation of pathways in recipient cells was determined at gene and protein expression levels. V-ATPase activity was impaired by Bafilomycin A1 or gene silencing. FINDINGS: GBM neurospheres influence their non-neoplastic microenvironment by delivering the V-ATPase subunit V1G1 and the homeobox genes HOXA7, HOXA10, and POU3F2 to recipient cells via LO. LOs reprogram recipient cells to proliferate, grow as spheres and to migrate. Moreover, LOs are particularly abundant in the circulation of GBM patients with short survival time. Finally, impairment of V-ATPase reduces LOs activity. INTERPRETATION: We identified a novel mechanism adopted by glioma stem cells to promote disease progression via LO-mediated reprogramming of their microenvironment. Our data provide preliminary evidence for future development of LO-based liquid biopsies and suggest a novel potential strategy to contrast glioma progression. FUND: This work was supported by Fondazione Cariplo (2014-1148 to VV) and by the Italian Minister of Health-Ricerca Corrente program 2017 (to SF).


Assuntos
Comunicação Autócrina , Neoplasias Encefálicas/metabolismo , Micropartículas Derivadas de Células/metabolismo , Glioblastoma/metabolismo , ATPases Vacuolares Próton-Translocadoras/metabolismo , Animais , Neoplasias Encefálicas/patologia , Linhagem Celular Tumoral , Células Cultivadas , Glioblastoma/patologia , Proteínas Homeobox A10 , Proteínas de Homeodomínio/metabolismo , Humanos , Camundongos , Fatores do Domínio POU/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Transdução de Sinais , Microambiente Tumoral , ATPases Vacuolares Próton-Translocadoras/genética
7.
EBioMedicine ; 41: 214-224, 2019 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-30737087

RESUMO

BACKGROUND: Cancer cells use specific V-ATPase subunits to activate oncogenic pathways. Therefore, we investigated V-ATPase deregulation in aggressive gliomas and associated signaling. METHODS: V-ATPase genes expression and associated pathways were analyzed in different series of glioma available from public databases, as well as in patients' cohort. Activation of pathways was analyzed at gene and protein expression levels. A genetic model of glioma in Drosophila melanogaster and mice with GBM patients-derived orthotopic xenografts were used as in vivo models of disease. FINDINGS: GBM and recurrent gliomas display a specific V-ATPase signature. Such signature resolves the heterogeneous class of IDH-wild type lower-grade gliomas, identifying the patients with worse prognosis independently from clinical and molecular features (p = 0·03, by Cox proportional-hazards model). In vivo, V-ATPase subunits deregulation significantly impacts tumor growth and proliferation. At the molecular level, GBM-like V-ATPase expression correlates with upregulation of Homeobox genes. INTERPRETATION: Our data identify a V-ATPase signature that accompanies glioma aggressiveness and suggest new entry points for glioma stratification and follow-up. FUND: This work was supported by Fondazione Cariplo (2014-1148 to VV), Fondazione IRCCS Ca' Granda, and Fondazione INGM Grant in Molecular Medicine 2014 (to VV).


Assuntos
Biomarcadores Tumorais/genética , Neoplasias Encefálicas/metabolismo , Glioma/metabolismo , ATPases Vacuolares Próton-Translocadoras/genética , Animais , Biomarcadores Tumorais/metabolismo , Neoplasias Encefálicas/classificação , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patologia , Células Cultivadas , Drosophila melanogaster , Feminino , Glioma/classificação , Glioma/genética , Glioma/patologia , Humanos , Isocitrato Desidrogenase/genética , Masculino , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , ATPases Vacuolares Próton-Translocadoras/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
8.
Autophagy ; 15(4): 631-651, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30335591

RESUMO

Macroautophagy/autophagy, a defense mechanism against aberrant stresses, in neurons counteracts aggregate-prone misfolded protein toxicity. Autophagy induction might be beneficial in neurodegenerative diseases (NDs). The natural compound trehalose promotes autophagy via TFEB (transcription factor EB), ameliorating disease phenotype in multiple ND models, but its mechanism is still obscure. We demonstrated that trehalose regulates autophagy by inducing rapid and transient lysosomal enlargement and membrane permeabilization (LMP). This effect correlated with the calcium-dependent phosphatase PPP3/calcineurin activation, TFEB dephosphorylation and nuclear translocation. Trehalose upregulated genes for the TFEB target and regulator Ppargc1a, lysosomal hydrolases and membrane proteins (Ctsb, Gla, Lamp2a, Mcoln1, Tpp1) and several autophagy-related components (Becn1, Atg10, Atg12, Sqstm1/p62, Map1lc3b, Hspb8 and Bag3) mostly in a PPP3- and TFEB-dependent manner. TFEB silencing counteracted the trehalose pro-degradative activity on misfolded protein causative of motoneuron diseases. Similar effects were exerted by trehalase-resistant trehalose analogs, melibiose and lactulose. Thus, limited lysosomal damage might induce autophagy, perhaps as a compensatory mechanism, a process that is beneficial to counteract neurodegeneration. Abbreviations: ALS: amyotrophic lateral sclerosis; AR: androgen receptor; ATG: autophagy related; AV: autophagic vacuole; BAG3: BCL2-associated athanogene 3; BECN1: beclin 1, autophagy related; CASA: chaperone-assisted selective autophagy; CTSB: cathepsin b; DAPI: 4',6-diamidino-2-phenylindole; DMEM: Dulbecco's modified Eagle's medium; EGFP: enhanced green fluorescent protein; fALS, familial amyotrophic lateral sclerosis; FRA: filter retardation assay; GAPDH: glyceraldehyde-3-phosphate dehydrogenase; GLA: galactosidase, alpha; HD: Huntington disease; hIPSCs: human induced pluripotent stem cells; HSPA8: heat shock protein A8; HSPB8: heat shock protein B8; IF: immunofluorescence analysis; LAMP1: lysosomal-associated membrane protein 1; LAMP2A: lysosomal-associated membrane protein 2A; LGALS3: lectin, galactose binding, soluble 3; LLOMe: L-leucyl-L-leucine methyl ester; LMP: lysosomal membrane permeabilization; Lys: lysosomes; MAP1LC3B: microtubule-associated protein 1 light chain 3 beta; MCOLN1: mucolipin 1; mRNA: messenger RNA; MTOR: mechanistic target of rapamycin kinase; NDs: neurodegenerative diseases; NSC34: neuroblastoma x spinal cord 34; PBS: phosphate-buffered saline; PD: Parkinson disease; polyQ: polyglutamine; PPARGC1A: peroxisome proliferative activated receptor, gamma, coactivator 1 alpha; PPP3CB: protein phosphatase 3, catalytic subunit, beta isoform; RT-qPCR: real-time quantitative polymerase chain reaction; SBMA: spinal and bulbar muscular atrophy; SCAs: spinocerebellar ataxias; siRNA: small interfering RNA; SLC2A8: solute carrier family 2, (facilitated glucose transporter), member 8; smNPCs: small molecules neural progenitors cells; SOD1: superoxide dismutase 1; SQSTM1/p62: sequestosome 1; STED: stimulated emission depletion; STUB1: STIP1 homology and U-box containing protein 1; TARDBP/TDP-43: TAR DNA binding protein; TFEB: transcription factor EB; TPP1: tripeptidyl peptidase I; TREH: trehalase (brush-border membrane glycoprotein); WB: western blotting; ZKSCAN3: zinc finger with KRAB and SCAN domains 3.


Assuntos
Autofagia/efeitos dos fármacos , Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos/metabolismo , Calcineurina/metabolismo , Lisossomos/metabolismo , Neurônios Motores/metabolismo , Neurônios Motores/patologia , Trealose/farmacologia , Esclerose Lateral Amiotrófica/tratamento farmacológico , Esclerose Lateral Amiotrófica/metabolismo , Animais , Autofagossomos/efeitos dos fármacos , Autofagossomos/enzimologia , Autofagossomos/metabolismo , Autofagia/genética , Proteínas Relacionadas à Autofagia/genética , Proteínas Relacionadas à Autofagia/metabolismo , Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos/química , Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos/genética , Atrofia Bulboespinal Ligada ao X/tratamento farmacológico , Atrofia Bulboespinal Ligada ao X/metabolismo , Calcineurina/genética , Cálcio/metabolismo , Diferenciação Celular , Núcleo Celular/metabolismo , Regulação para Baixo/genética , Humanos , Células-Tronco Pluripotentes Induzidas/enzimologia , Células-Tronco Pluripotentes Induzidas/metabolismo , Células-Tronco Pluripotentes Induzidas/patologia , Células-Tronco Pluripotentes Induzidas/ultraestrutura , Lisossomos/efeitos dos fármacos , Lisossomos/enzimologia , Lisossomos/ultraestrutura , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Camundongos , Proteínas Associadas aos Microtúbulos/metabolismo , Neurônios Motores/enzimologia , Neurônios Motores/ultraestrutura , Neuroproteção/efeitos dos fármacos , Neuroproteção/genética , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Proteína Sequestossoma-1/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Trealose/análogos & derivados , Tripeptidil-Peptidase 1 , Resposta a Proteínas não Dobradas/genética
9.
Swiss Med Wkly ; 146: w14347, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27631343

RESUMO

Originally discovered as regulators of cargo sorting during endosomal trafficking, ESCRT (endosomal sorting complexes required for transport) proteins are emerging as flexible machines that shape the behaviour of membranes throughout the cell. Deregulation of ESCRT activity is observed in neuro-degenerative diseases, virus infection and cancer. However, the mechanisms of pathogenesis involving ESCRTs have not yet fully come into focus. Here, we review the current knowledge of ESCRT function in health and disease and provide educated guesses for future research and focused therapeutic intervention.


Assuntos
Complexos Endossomais de Distribuição Requeridos para Transporte , Animais , Complexos Endossomais de Distribuição Requeridos para Transporte/genética , Complexos Endossomais de Distribuição Requeridos para Transporte/fisiologia , Complexos Endossomais de Distribuição Requeridos para Transporte/uso terapêutico , Endossomos , Humanos , Mutação , Neoplasias/fisiopatologia , Neoplasias/terapia , Doenças Neurodegenerativas/fisiopatologia , Doenças Neurodegenerativas/terapia , Membrana Nuclear , Viroses/fisiopatologia , Viroses/terapia
10.
Autophagy ; 12(3): 499-514, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26727288

RESUMO

In vertebrates, TFEB (transcription factor EB) and MITF (microphthalmia-associated transcription factor) family of basic Helix-Loop-Helix (bHLH) transcription factors regulates both lysosomal function and organ development. However, it is not clear whether these 2 processes are interconnected. Here, we show that Mitf, the single TFEB and MITF ortholog in Drosophila, controls expression of vacuolar-type H(+)-ATPase pump (V-ATPase) subunits. Remarkably, we also find that expression of Vha16-1 and Vha13, encoding 2 key components of V-ATPase, is patterned in the wing imaginal disc. In particular, Vha16-1 expression follows differentiation of proneural regions of the disc. These regions, which will form sensory organs in the adult, appear to possess a distinctive endolysosomal compartment and Notch (N) localization. Modulation of Mitf activity in the disc in vivo alters endolysosomal function and disrupts proneural patterning. Similar to our findings in Drosophila, in human breast epithelial cells we observe that impairment of the Vha16-1 human ortholog ATP6V0C changes the size and function of the endolysosomal compartment and that depletion of TFEB reduces ligand-independent N signaling activity. Our data suggest that lysosomal-associated functions regulated by the TFEB-V-ATPase axis might play a conserved role in shaping cell fate.


Assuntos
Padronização Corporal , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Lisossomos/metabolismo , Receptores Notch/metabolismo , Transdução de Sinais , ATPases Vacuolares Próton-Translocadoras/metabolismo , Animais , Diferenciação Celular , Drosophila melanogaster/citologia , Drosophila melanogaster/genética , Células Epiteliais/metabolismo , Humanos , Discos Imaginais/metabolismo , Modelos Biológicos , Neurônios/citologia , Neurônios/metabolismo , Homologia de Sequência de Aminoácidos , Transcrição Gênica , ATPases Vacuolares Próton-Translocadoras/genética , Asas de Animais/metabolismo
11.
Oncotarget ; 6(19): 17514-31, 2015 Jul 10.
Artigo em Inglês | MEDLINE | ID: mdl-26020805

RESUMO

The vacuolar H+ ATPase (V-ATPase) is a proton pump responsible for acidification of cellular microenvironments, an activity exploited by tumors to survive, proliferate and resist to therapy. Despite few observations, the role of V-ATPase in human tumorigenesis remains unclear.We investigated the expression of ATP6V0C, ATP6V0A2, encoding two subunits belonging to the V-ATPase V0 sector and ATP6V1C, ATP6V1G1, ATPT6V1G2, ATP6V1G3, which are part of the V1 sector, in series of adult gliomas and in cancer stem cell-enriched neurospheres isolated from glioblastoma (GBM) patients. ATP6V1G1 expression resulted significantly upregulated in tissues of patients with GBM and correlated with shorter patients' overall survival independent of clinical variables.ATP6V1G1 knockdown in GBM neurospheres hampered sphere-forming ability, induced cell death, and decreased matrix invasion, a phenotype not observed in GBM monolayer cultures. Treating GBM organotypic cultures or neurospheres with the selective V-ATPase inhibitor bafilomycin A1 reproduced the effects of ATP6V1G1 siRNA and strongly suppressed expression of the stem cell markers Nestin, CD133 and transcription factors SALL2 and POU3F2 in neurospheres.These data point to ATP6V1G1 as a novel marker of poor prognosis in GBM patients and identify V-ATPase inhibition as an innovative therapeutic strategy for GBM.


Assuntos
Neoplasias Encefálicas/patologia , Glioblastoma/patologia , ATPases Vacuolares Próton-Translocadoras/metabolismo , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Neoplasias Encefálicas/enzimologia , Neoplasias Encefálicas/mortalidade , Movimento Celular , Sobrevivência Celular , Feminino , Imunofluorescência , Glioblastoma/enzimologia , Glioblastoma/mortalidade , Humanos , Estimativa de Kaplan-Meier , Masculino , Pessoa de Meia-Idade , Prognóstico , RNA Interferente Pequeno , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Análise Serial de Tecidos , Transfecção , Adulto Jovem
12.
PLoS One ; 9(4): e93987, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24718108

RESUMO

Multivesicular endosome (MVE) sorting depends on proteins of the Endosomal Sorting Complex Required for Transport (ESCRT) family. These are organized in four complexes (ESCRT-0, -I, -II, -III) that act in a sequential fashion to deliver ubiquitylated cargoes into the internal luminal vesicles (ILVs) of the MVE. Drosophila genes encoding ESCRT-I, -II, -III components function in sorting signaling receptors, including Notch and the JAK/STAT signaling receptor Domeless. Loss of ESCRT-I, -II, -III in Drosophila epithelia causes altered signaling and cell polarity, suggesting that ESCRTs genes are tumor suppressors. However, the nature of the tumor suppressive function of ESCRTs, and whether tumor suppression is linked to receptor sorting is unclear. Unexpectedly, a null mutant in Hrs, encoding one of the components of the ESCRT-0 complex, which acts upstream of ESCRT-I, -II, -III in MVE sorting is dispensable for tumor suppression. Here, we report that two Drosophila epithelia lacking activity of Stam, the other known components of the ESCRT-0 complex, or of both Hrs and Stam, accumulate the signaling receptors Notch and Dome in endosomes. However, mutant tissue surprisingly maintains normal apico-basal polarity and proliferation control and does not display ectopic Notch signaling activation, unlike cells that lack ESCRT-I, -II, -III activity. Overall, our in vivo data confirm previous evidence indicating that the ESCRT-0 complex plays no crucial role in regulation of tumor suppression, and suggest re-evaluation of the relationship of signaling modulation in endosomes and tumorigenesis.


Assuntos
Proteínas de Drosophila/deficiência , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/fisiologia , Complexos Endossomais de Distribuição Requeridos para Transporte/fisiologia , Endossomos/fisiologia , Células Epiteliais/metabolismo , Genes Supressores de Tumor , Receptores de Interleucina/metabolismo , Receptores Notch/metabolismo , Proteínas Supressoras de Tumor/deficiência , Proteínas Adaptadoras de Transdução de Sinal/deficiência , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transdução de Sinal/fisiologia , Animais , Transformação Celular Neoplásica/genética , Deleção Cromossômica , Cromossomos de Insetos/genética , Proteínas de Drosophila/genética , Proteínas de Drosophila/fisiologia , Drosophila melanogaster/genética , Complexos Endossomais de Distribuição Requeridos para Transporte/deficiência , Complexos Endossomais de Distribuição Requeridos para Transporte/genética , Endossomos/ultraestrutura , Olho/crescimento & desenvolvimento , Olho/patologia , Feminino , Teste de Complementação Genética , Discos Imaginais/metabolismo , Discos Imaginais/patologia , Mosaicismo , Folículo Ovariano/patologia , Fosfoproteínas/deficiência , Fosfoproteínas/genética , Fosfoproteínas/fisiologia , Transporte Proteico/fisiologia , Transdução de Sinais/genética , Transdução de Sinais/fisiologia , Proteínas Supressoras de Tumor/genética
13.
Mol Oncol ; 8(2): 207-20, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24309677

RESUMO

Notch signaling in prominently involved in growth regulation in metazoan tissues. Because of this, Notch is often upregulated in cancer and current efforts point to developing drugs that block its activation. Notch receptor endocytosis towards acidic compartments is a recently appreciated determinant of signaling activation. Vacuolar H(+) ATPase (V-ATPase) is responsible for acidification of endocytic organelles and mutants in V-ATPase subunit encoding genes in model organisms have been recently shown to display loss of Notch signaling. Here, we show that administration of BafilomycinA1 (BafA1), a highly specific V-ATPase inhibitor decreases Notch signaling during Drosophila and Zebrafish development, and in human cells in culture. In normal breast cells, we find that BafA1 treatment leads to accumulation of Notch in the endo-lysosomal system, and reduces its processing and signaling activity. In Notch-addicted breast cancer cells, BafA1 treatment reduces growth in cells expressing membrane tethered forms of Notch, while sparing cells expressing cytoplasmic forms. In contrast, we find that V-ATPase inhibition reduces growth of leukemia cells, without affecting Notch activatory cleavage. However, consistent with the emerging roles of V-ATPase in controlling multiple signaling pathways, in these cells Akt activation is reduced, as it is also the case in BafA1-treated breast cancer cells. Our data support V-ATPase inhibition as a novel therapeutic approach to counteract tumor growth via signaling pathways regulated at the endo-lysosomal level.


Assuntos
Neoplasias da Mama/metabolismo , Proteínas de Drosophila/antagonistas & inibidores , Inibidores Enzimáticos/farmacologia , Macrolídeos/farmacologia , Proteínas de Neoplasias/antagonistas & inibidores , Receptores Notch/antagonistas & inibidores , Transdução de Sinais , ATPases Vacuolares Próton-Translocadoras/metabolismo , Proteínas de Peixe-Zebra/metabolismo , Peixe-Zebra/metabolismo , Animais , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster , Feminino , Humanos , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Receptores Notch/genética , Receptores Notch/metabolismo , ATPases Vacuolares Próton-Translocadoras/genética , Peixe-Zebra/genética , Proteínas de Peixe-Zebra/genética
14.
Dis Model Mech ; 6(3): 689-700, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23335205

RESUMO

The C subunit of the vacuolar H(+)-ATPase or V-ATPase regulates the activity and assembly of the proton pump at cellular membranes. It has been shown to be strongly upregulated in oral squamous cell carcinoma, a highly metastatic epithelial cancer. In addition, increased V-ATPase activity appears to correlate with invasiveness of cancer cells, but the underlying mechanism is largely unknown. Using the Drosophila wing imaginal epithelium as an in vivo model system, we demonstrate that overexpression of Vha44, the Drosophila orthologue of the C subunit, causes a tumor-like tissue transformation in cells of the wing epithelium. Overexpressing cells are excluded from the epithelium and acquire invasive properties while displaying high apoptotic rates. Blocking apoptosis in these cells unmasks a strong proliferation stimulus, leading to overgrowth. Furthermore, we show that excess Vha44 greatly increases acidification of endocytic compartments and interferes with endosomal trafficking. As a result, cargoes such as GFP-Lamp1 and Notch accumulate in highly acidified enlarged endolysosomal compartments. Consistent with previous reports on the endocytic activation of Eiger/JNK signaling, we find that V-ATPase stimulation by Vha44 causes JNK signaling activation whereas downmodulation of JNK signaling rescues the invasive phenotypes. In summary, our in vivo-findings demonstrate that increased levels of V-ATPase C subunit induce a Eiger/JNK-dependent cell transformation within an epithelial organ that recapitulates early carcinoma stages.


Assuntos
Movimento Celular , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/enzimologia , Drosophila melanogaster/crescimento & desenvolvimento , Epitélio/crescimento & desenvolvimento , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Subunidades Proteicas/metabolismo , ATPases Vacuolares Próton-Translocadoras/metabolismo , Animais , Apoptose , Padronização Corporal , Compartimento Celular , Drosophila melanogaster/citologia , Endossomos/metabolismo , Epitélio/enzimologia , Concentração de Íons de Hidrogênio , Lisossomos/metabolismo , Sistema de Sinalização das MAP Quinases , Proteínas de Membrana/metabolismo , Modelos Biológicos , Proteínas Mutantes/metabolismo , Oncogenes , Proteólise , Receptores Notch/metabolismo , Asas de Animais/citologia , Asas de Animais/crescimento & desenvolvimento , Proteínas ras/metabolismo
15.
Nat Genet ; 41(10): 1150-5, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19749759

RESUMO

A prevailing paradigm posits that Polycomb Group (PcG) proteins maintain stem cell identity by repressing differentiation genes, and abundant evidence points to an oncogenic role for PcG proteins in human cancer. Here we show using Drosophila melanogaster that a conventional PcG complex can also have a potent tumor suppressor activity. Mutations in any core PRC1 component cause pronounced hyperproliferation of eye imaginal tissue, accompanied by deregulation of epithelial architecture. The mitogenic JAK-STAT pathway is strongly and specifically activated in mutant tissue; activation is driven by transcriptional upregulation of Unpaired (Upd, also known as Outstretched, Os) family ligands. We show here that upd genes are direct targets of PcG-mediated repression in imaginal discs. Ectopic JAK-STAT activity is sufficient to induce overproliferation, whereas reduction of JAK-STAT activity suppresses the PRC1 mutant tumor phenotype. These findings show that PcG proteins can restrict growth directly by silencing mitogenic signaling pathways, shedding light on an epigenetic mechanism underlying tumor suppression.


Assuntos
Drosophila melanogaster/metabolismo , Janus Quinases/metabolismo , Proteínas Repressoras/metabolismo , Fatores de Transcrição STAT/metabolismo , Transdução de Sinais , Animais , Proliferação de Células , Drosophila melanogaster/citologia , Drosophila melanogaster/genética , Olho/citologia , Olho/metabolismo , Neoplasias/genética , Neoplasias/metabolismo , Neoplasias/patologia , Proteínas do Grupo Polycomb , Proteínas Repressoras/genética
16.
Mol Oncol ; 3(4): 354-65, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19560990

RESUMO

Endocytosis is an important regulator of cell-cell signaling and endocytic trafficking has been increasingly implicated in control of tumor suppression. Recent insights from Drosophila indicate that impairment of multiple trafficking steps which lead to receptor degradation can cause tumor formation in epithelial organs. These tumors are characterized by sustained activation of a number of mitogenic signaling pathways, and by subversion of epithelial polarity and the apoptotic response. Cooperation between such alterations, as well as tumor-host interactions, is also observed. The recapitulation of several hallmarks of human cancers in fly tumors provides a framework to understand the role of defective endocytosis in cancer.


Assuntos
Apoptose/fisiologia , Polaridade Celular/fisiologia , Proliferação de Células , Drosophila/fisiologia , Endocitose/fisiologia , Neoplasias/patologia , Animais , Apoptose/genética , Polaridade Celular/genética , Drosophila/citologia , Endocitose/genética , Humanos , Modelos Biológicos , Neoplasias/metabolismo
17.
Genetics ; 177(3): 1667-77, 2007 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17947427

RESUMO

The Drosophila neoplastic tumor suppressor genes (TSGs) coordinately control cell polarity and proliferation in epithelial and neuronal tissues. While a small group of neoplastic TSG mutations have been isolated and their corresponding genes cloned, the regulatory pathways that normally prevent inappropriate growth remain unclear. Identification of additional neoplastic TSGs may provide insight into this question. We report here the design of an efficient screen for isolating neoplastic TSG mutations utilizing genetically mosaic larvae. This screen is based on a defective pupation phenotype seen when a single pair of imaginal discs is homozygous for a neoplastic TSG mutation, which suggests that continuously proliferating cells can interfere with metamorphosis. Execution of this screen on two chromosome arms led to the identification of mutations in at least seven new neoplastic TSGs. The isolation of additional loci that affect hyperplastic as well as neoplastic growth indicates the utility of this screening strategy for studying epithelial growth control.


Assuntos
Drosophila/genética , Genes de Insetos , Genes Supressores de Tumor , Animais , Drosophila/crescimento & desenvolvimento , Olho/crescimento & desenvolvimento , Feminino , Genes Letais , Teste de Complementação Genética , Masculino , Metamorfose Biológica , Mosaicismo , Mutação , Fenótipo , Pupa/crescimento & desenvolvimento
18.
Dev Cell ; 9(5): 687-98, 2005 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-16256743

RESUMO

Cell-cell signaling coordinates proliferation of metazoan tissues during development, and its alteration can induce malignant transformation. Endocytosis regulates signaling by controlling the levels and activity of transmembrane receptors, both prior to and following ligand engagement. Here, we identify Vps25, a component of the ESCRT machinery that regulates endocytic sorting of signaling receptors, as an unconventional type of Drosophila tumor suppressor. vps25 mutant cells undergo autonomous neoplastic-like transformation, but they also stimulate nonautonomous cell proliferation. Endocytic trafficking defects in vps25 cells cause endosomal accumulation of the signaling receptor Notch and enhanced Notch signaling. Increased Notch activity leads to ectopic production of the mitogenic JAK-STAT pathway ligand Unpaired, which is secreted from mutant cells to induce overproliferation of the surrounding epithelium. Our data show that defects in endocytic sorting can both transform cells and, through heterotypic signaling, alter the behavior of neighboring wild-type tissue.


Assuntos
Proteínas de Drosophila/metabolismo , Drosophila/genética , Receptores Notch/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Animais , Proliferação de Células , Drosophila/metabolismo , Proteínas de Drosophila/genética , Endossomos/genética , Endossomos/metabolismo , Regulação da Expressão Gênica/genética , Mutação , Transporte Proteico/fisiologia , Receptores Notch/genética , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Proteínas Supressoras de Tumor/genética
19.
Curr Biol ; 12(17): 1524-8, 2002 Sep 03.
Artigo em Inglês | MEDLINE | ID: mdl-12225669

RESUMO

After its specification, the Drosophila oocyte undergoes a critical polarization event that involves a reorganization of the microtubules (MT) and relocalization of the determinant Orb within the oocyte. This polarization requires Par-1 kinase and the PDZ-containing Par-3 homolog, Bazooka (Baz). Par-1 has been observed on the fusome, which degenerates before the onset of oocyte polarization. How Par-1 acts to polarize the oocyte has been unclear. Here we show that Par-1 becomes restricted to the oocyte in a MT-dependent fashion after disappearance of the fusome. At the time of polarization, the kinase itself and the determinant BicaudalD (BicD) are relocalized from the anterior to the posterior of the oocyte. Par-1 and BicD are interdependent and require MT and the minus end-directed motor Dynein for their relocalization. We show that baz is required for Par-1 relocalization within the oocyte and that the distributions of Baz and Par-1 in the Drosophila oocyte are complementary and strikingly reminiscent of the two PAR proteins in the C. elegans embryo. We propose that, through the combined actions of the fusome, MT, and Baz, Par-1 is selectively enriched and localized within the oocyte, where, in conjunction with BicD, Egalitarian (Egl), and Dynein, it acts on the MT cytoskeleton to effect polarization.


Assuntos
Proteínas de Transporte/fisiologia , Polaridade Celular/fisiologia , Citoesqueleto/ultraestrutura , Proteínas de Drosophila/fisiologia , Drosophila melanogaster/fisiologia , Dineínas/fisiologia , Proteínas do Ovo/fisiologia , Peptídeos e Proteínas de Sinalização Intracelular , Microtúbulos/fisiologia , Oócitos/ultraestrutura , Organelas/ultraestrutura , Proteínas Quinases/fisiologia , Animais , Proteínas de Transporte/genética , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/citologia , Drosophila melanogaster/genética , Dineínas/genética , Proteínas do Ovo/genética , Quinase 3 da Glicogênio Sintase , Proteínas Motores Moleculares , Oócitos/metabolismo , Oogênese , Proteínas Quinases/genética , Proteínas Serina-Treonina Quinases , Proteínas de Ligação a RNA/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA