Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 4 de 4
Filtrar
Mais filtros

Bases de dados
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
J Biol Chem ; 293(25): 9784-9800, 2018 06 22.
Artigo em Inglês | MEDLINE | ID: mdl-29748384

RESUMO

Telomerase activation protects cells from telomere damage by delaying senescence and inducing cell immortalization, whereas telomerase inhibition mediates rapid senescence or apoptosis. However, the cellular mechanisms that determine telomere damage-dependent senescence versus apoptosis induction are largely unknown. Here, we demonstrate that telomerase instability mediated by silencing of sphingosine kinase 2 (SPHK2) and sphingosine 1-phosphate (S1P), which binds and stabilizes telomerase, induces telomere damage-dependent caspase-3 activation and apoptosis, but not senescence, in p16-deficient lung cancer cells or tumors. These outcomes were prevented by knockdown of a tumor-suppressor protein, transcription factor 21 (TCF21), or by ectopic expression of WT human telomerase reverse transcriptase (hTERT) but not mutant hTERT with altered S1P binding. Interestingly, SphK2-deficient mice exhibited accelerated aging and telomerase instability that increased telomere damage and senescence via p16 activation especially in testes tissues, but not in apoptosis. Moreover, p16 silencing in SphK2-/- mouse embryonic fibroblasts activated caspase-3 and apoptosis without inducing senescence. Furthermore, ectopic WT p16 expression in p16-deficient A549 lung cancer cells prevented TCF21 and caspase-3 activation and resulted in senescence in response to SphK2/S1P inhibition and telomere damage. Mechanistically, a p16 mutant with impaired caspase-3 association did not prevent telomere damage-induced apoptosis, indicating that an association between p16 and caspase-3 proteins forces senescence induction by inhibiting caspase-3 activation and apoptosis. These results suggest that p16 plays a direct role in telomere damage-dependent senescence by limiting apoptosis via binding to caspase-3, revealing a direct link between telomere damage-dependent senescence and apoptosis with regards to aging and cancer.


Assuntos
Apoptose , Caspase 3/metabolismo , Senescência Celular , Inibidor p16 de Quinase Dependente de Ciclina/metabolismo , Neoplasias Pulmonares/patologia , Fosfotransferases (Aceptor do Grupo Álcool)/fisiologia , Telômero/patologia , Animais , Caspase 3/genética , Proliferação de Células , Inibidor p16 de Quinase Dependente de Ciclina/genética , Feminino , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Lisofosfolipídeos/metabolismo , Camundongos , Camundongos Knockout , Camundongos SCID , Fosfotransferases (Aceptor do Grupo Álcool)/genética , Fosfotransferases (Aceptor do Grupo Álcool)/metabolismo , Esfingosina/análogos & derivados , Esfingosina/metabolismo , Telômero/genética , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
2.
Sci Signal ; 10(502)2017 Oct 24.
Artigo em Inglês | MEDLINE | ID: mdl-29066540

RESUMO

Signaling by the transforming growth factor-ß (TGF-ß) receptors I and II (TßRI/II) and the primary cilia-localized sonic hedgehog (Shh) pathway promote cell migration and, consequently, tumor metastasis. In contrast, the sphingolipid ceramide inhibits cell proliferation and tumor metastasis. We investigated whether ceramide metabolism inhibited TßRI/II trafficking to primary cilia to attenuate cross-talk between TßRI/II and the Shh pathway. We found that ceramide synthase 4 (CerS4)-generated ceramide stabilized the association between TßRI and the inhibitory factor Smad7, which limited the trafficking of TßRI/II to primary cilia. Expression of a mutant TßRI that signals but does not interact with Smad7 prevented the CerS4-mediated inhibition of migration in various cancer cells. Genetic deletion or knockdown of CerS4 prevented the formation of the Smad7-TßRI inhibitory complex and increased the association between TßRI and the transporter Arl6 through a previously unknown cilia-targeting signal (Ala31Thr32Ala33Leu34Gln35) in TßRI. Mutating the cilia-targeting signal abolished the trafficking of TßRI to the primary cilia. Localization of TßRI to primary cilia activated a key mediator of Shh signaling, Smoothened (Smo), which stimulated cellular migration and invasion. TßRI-Smo cross-talk at the cilia in CerS4-deficient 4T1 mammary cancer cells induced liver metastasis from orthotopic allografts in both wild-type and CerS4-deficient mice, which was prevented by overexpression of Smad7 or knockdown of intraflagellar transport protein 88 (IFT88). Overall, these data reveal a ceramide-dependent mechanism that suppresses cell migration and invasion by restricting TßRI/II-Shh signaling selectively at the plasma membrane of the primary cilium.


Assuntos
Movimento Celular , Ceramidas/metabolismo , Cílios/metabolismo , Metástase Neoplásica/patologia , Receptores de Fatores de Crescimento Transformadores beta/metabolismo , Células A549 , Animais , Linhagem Celular Tumoral , Proliferação de Células , Ceramidas/genética , Cílios/genética , Modelos Animais de Doenças , Técnicas de Silenciamento de Genes , Humanos , Camundongos , Camundongos Knockout , Receptores de Fatores de Crescimento Transformadores beta/genética , Proteína Smad7/metabolismo , Esfingosina N-Aciltransferase/genética , Esfingosina N-Aciltransferase/metabolismo
3.
EMBO Mol Med ; 9(8): 1030-1051, 2017 08.
Artigo em Inglês | MEDLINE | ID: mdl-28606997

RESUMO

Human papillomavirus (HPV) infection is linked to improved survival in response to chemo-radiotherapy for patients with oropharynx head and neck squamous cell carcinoma (HNSCC). However, mechanisms involved in increased HNSCC cell death by HPV signaling in response to therapy are largely unknown. Here, using molecular, pharmacologic and genetic tools, we show that HPV early protein 7 (E7) enhances ceramide-mediated lethal mitophagy in response to chemotherapy-induced cellular stress in HPV-positive HNSCC cells by selectively targeting retinoblastoma protein (RB). Inhibition of RB by HPV-E7 relieves E2F5, which then associates with DRP1, providing a scaffolding platform for Drp1 activation and mitochondrial translocation, leading to mitochondrial fission and increased lethal mitophagy. Ectopic expression of a constitutively active mutant RB, which is not inhibited by HPV-E7, attenuated ceramide-dependent mitophagy and cell death in HPV(+) HNSCC cells. Moreover, mutation of E2F5 to prevent Drp1 activation inhibited mitophagy in HPV(+) cells. Activation of Drp1 with E2F5-mimetic peptide for inducing Drp1 mitochondrial localization enhanced ceramide-mediated mitophagy and led to tumor suppression in HPV-negative HNSCC-derived xenograft tumors in response to cisplatin in SCID mice.


Assuntos
Antineoplásicos/administração & dosagem , Carcinoma de Células Escamosas/tratamento farmacológico , Ceramidas/metabolismo , Cisplatino/administração & dosagem , Proteínas de Membrana/metabolismo , Mitofagia , Proteínas E7 de Papillomavirus/metabolismo , Esfingosina N-Aciltransferase/metabolismo , Animais , Antineoplásicos/metabolismo , Carcinoma de Células Escamosas/patologia , Morte Celular , Linhagem Celular Tumoral , Cisplatino/metabolismo , Modelos Animais de Doenças , Xenoenxertos , Humanos , Camundongos SCID , Transplante de Neoplasias , Proteínas E7 de Papillomavirus/genética , Resultado do Tratamento
4.
Sci Signal ; 8(381): ra58, 2015 Jun 16.
Artigo em Inglês | MEDLINE | ID: mdl-26082434

RESUMO

During DNA replication, the enzyme telomerase maintains the ends of chromosomes, called telomeres. Shortened telomeres trigger cell senescence, and cancer cells often have increased telomerase activity to promote their ability to proliferate indefinitely. The catalytic subunit, human telomerase reverse transcriptase (hTERT), is stabilized by phosphorylation. We found that the lysophospholipid sphingosine 1-phosphate (S1P), generated by sphingosine kinase 2 (SK2), bound hTERT at the nuclear periphery in human and mouse fibroblasts. Docking predictions and mutational analyses revealed that binding occurred between a hydroxyl group (C'3-OH) in S1P and Asp(684) in hTERT. Inhibiting or depleting SK2 or mutating the S1P binding site decreased the stability of hTERT in cultured cells and promoted senescence and loss of telomere integrity. S1P binding inhibited the interaction of hTERT with makorin ring finger protein 1 (MKRN1), an E3 ubiquitin ligase that tags hTERT for degradation. Murine Lewis lung carcinoma (LLC) cells formed smaller tumors in mice lacking SK2 than in wild-type mice, and knocking down SK2 in LLC cells before implantation into mice suppressed their growth. Pharmacologically inhibiting SK2 decreased the growth of subcutaneous A549 lung cancer cell-derived xenografts in mice, and expression of wild-type hTERT, but not an S1P-binding mutant, restored tumor growth. Thus, our data suggest that S1P binding to hTERT allosterically mimicks phosphorylation, promoting telomerase stability and hence telomere maintenance, cell proliferation, and tumor growth.


Assuntos
Carcinoma Pulmonar de Lewis/metabolismo , Núcleo Celular/metabolismo , Lisofosfolipídeos/metabolismo , Simulação de Acoplamento Molecular , Proteínas de Neoplasias/metabolismo , Esfingosina/análogos & derivados , Telomerase/metabolismo , Regulação Alostérica/genética , Animais , Carcinoma Pulmonar de Lewis/genética , Linhagem Celular Tumoral , Núcleo Celular/genética , Núcleo Celular/patologia , Humanos , Lisofosfolipídeos/genética , Camundongos , Camundongos Knockout , Camundongos SCID , Proteínas de Neoplasias/antagonistas & inibidores , Proteínas de Neoplasias/genética , Fosforilação/genética , Ligação Proteica , Esfingosina/genética , Esfingosina/metabolismo , Telomerase/antagonistas & inibidores , Telomerase/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA