Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 2.570
Filtrar
1.
Nat Commun ; 13(1): 7579, 2022 12 08.
Artigo em Inglês | MEDLINE | ID: mdl-36482070

RESUMO

The adult brain retains over life endogenous neural stem/precursor cells (eNPCs) within the subventricular zone (SVZ). Whether or not these cells exert physiological functions is still unclear. In the present work, we provide evidence that SVZ-eNPCs tune structural, electrophysiological, and behavioural aspects of striatal function via secretion of insulin-like growth factor binding protein-like 1 (IGFBPL1). In mice, selective ablation of SVZ-eNPCs or selective abrogation of IGFBPL1 determined an impairment of striatal medium spiny neuron morphology, a higher failure rate in GABAergic transmission mediated by fast-spiking interneurons, and striatum-related behavioural dysfunctions. We also found IGFBPL1 expression in the human SVZ, foetal and induced-pluripotent stem cell-derived NPCs. Finally, we found a significant correlation between SVZ damage, reduction of striatum volume, and impairment of information processing speed in neurological patients. Our results highlight the physiological role of adult SVZ-eNPCs in supporting cognitive functions by regulating striatal neuronal activity.


Assuntos
Proteínas de Ligação a Fator de Crescimento Semelhante a Insulina , Ventrículos Laterais , Células-Tronco Neurais , Proteínas Supressoras de Tumor , Animais , Humanos , Camundongos , Eletrofisiologia Cardíaca , Proteínas de Ligação a Fator de Crescimento Semelhante a Insulina/fisiologia , Células-Tronco Neurais/fisiologia , Proteínas Supressoras de Tumor/fisiologia , Ventrículos Laterais/fisiologia
2.
Exp Eye Res ; 216: 108955, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-35074341

RESUMO

Krüppel-like factor 2 (KLF2) belongs to the KLF family of zinc-finger transcription factors and mediates the occurrence and progression of various cancers. However, little is known about its expression pattern and biological role in retinoblastoma (RB). In the present study, we showed that KLF2 was markedly downregulated in human RB tissue compared with retina. KLF2 overexpression significantly inhibited RB cell proliferation and decreased proliferating cell nuclear antigen (PCNA) expression. Subsequently, we confirmed that KLF2 arrested cells at the G1-S phase transition, accompanied by the upregulation of p21 and downregulation of CyclinD1, as well as the activation of mitochondria-mediated apoptosis in RB cells. In addition, KLF2 overexpression contributed to suppressing RB cell migration and invasion by downregulating matrix metallopeptidase 9 (MMP9). On the contrary, KLF2 downregulation promoted RB cells proliferation, migration and invasion. Notably, the KLF2 expression pattern was opposite to that of C-X-C chemokine receptor 4 (CXCR4) in the two RB cell lines, KLF2 overexpression significantly decreased CXCR4 expression, silencing KLF2 had the opposite effect. Furthermore, dual-luciferase reporter and chromatin immunoprecipitation (ChIP) assays confirmed that KLF2 directly bound to the CXCR4 promoter and negatively regulated its expression in RB cells. Collectively, our results suggested that KLF2 function as a tumor suppressor in RB and may represent a potential therapeutic target for RB.


Assuntos
Fatores de Transcrição Kruppel-Like/fisiologia , Neoplasias da Retina/metabolismo , Retinoblastoma/metabolismo , Proteínas Supressoras de Tumor/fisiologia , Apoptose/fisiologia , Western Blotting , Ciclo Celular/fisiologia , Linhagem Celular Tumoral , Proliferação de Células/fisiologia , Ciclina D1/genética , Regulação Neoplásica da Expressão Gênica/fisiologia , Humanos , Marcação In Situ das Extremidades Cortadas , Plasmídeos , Antígeno Nuclear de Célula em Proliferação/genética , RNA Interferente Pequeno/genética , Reação em Cadeia da Polimerase em Tempo Real , Neoplasias da Retina/patologia , Retinoblastoma/patologia , Transfecção , Quinases Ativadas por p21/genética
3.
Arthritis Rheumatol ; 74(1): 49-59, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34105311

RESUMO

OBJECTIVE: Mechanical and biologic cues drive cellular signaling in cartilage development, health, and disease. Primary cilia proteins, which are implicated in the transduction of biologic and physiochemical signals, control cartilage formation during skeletal development. This study was undertaken to assess the influence of the ciliary protein intraflagellar transport protein 88 (IFT88) on postnatal cartilage from mice with conditional knockout of the Ift88 gene (Ift88-KO). METHODS: Ift88fl/fl and aggrecanCreERT2 mice were crossed to create a strain of cartilage-specific Ift88-KO mice (aggrecanCreERT2 ;Ift88fl/fl ). In these Ift88-KO mice and Ift88fl/fl control mice, tibial articular cartilage thickness was assessed by histomorphometry, and the integrity of the cartilage was assessed using Osteoarthritis Research Society International (OARSI) damage scores, from adolescence through adulthood. In situ mechanisms of cartilage damage were investigated in the microdissected cartilage sections using immunohistochemistry, RNAScope analysis, and quantitative polymerase chain reaction. Osteoarthritis (OA) was induced in aggrecanCreERT2 ;Ift88fl/fl mice and Ift88fl/fl control mice using surgical destabilization of the medial meniscus (DMM). Following tamoxifen injection and DMM surgery, the mice were given free access to exercise on a wheel. RESULTS: Deletion of Ift88 resulted in progressive reduction in the thickness of the medial tibial cartilage in adolescent mice, as well as marked atrophy of the cartilage in mice during adulthood. In aggrecanCreERT2 ;Ift88fl/fl mice at age 34 weeks, the median thickness of the medial tibial cartilage was 89.42 µm (95% confidence interval [95% CI] 84.00-93.49), whereas in Ift88fl/fl controls at the same age, the median cartilage thickness was 104.00 µm (95% CI 100.30-110.50; P < 0.0001). At all time points, the median thickness of the calcified cartilage was reduced. In some mice, atrophy of the medial tibial cartilage was associated with complete, spontaneous degradation of the cartilage. Following DMM, aggrecanCreERT2 ;Ift88fl/fl mice were found to have increased OARSI scores of cartilage damage. In articular cartilage from maturing mice, atrophy was not associated with obvious increases in aggrecanase-mediated destruction or chondrocyte hypertrophy. Of the 44 candidate genes analyzed, only Tcf7l2 expression levels correlated with Ift88 expression levels in the microdissected cartilage. However, RNAScope analysis revealed that increased hedgehog (Hh) signaling (as indicated by increased expression of Gli1) was associated with the reductions in Ift88 expression in the tibial cartilage from Ift88-deficient mice. Wheel exercise restored both the articular cartilage thickness and levels of Hh signaling in these mice. CONCLUSION: Our results in a mouse model of OA demonstrate that IFT88 performs a chondroprotective role in articular cartilage by controlling the calcification of cartilage via maintenance of a threshold of Hh signaling during physiologic loading.


Assuntos
Cartilagem Articular/crescimento & desenvolvimento , Osteoartrite/etiologia , Proteínas Supressoras de Tumor/fisiologia , Animais , Cartilagem Articular/anatomia & histologia , Masculino , Camundongos , Camundongos Knockout , Tamanho do Órgão
4.
J Clin Invest ; 131(22)2021 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-34779410

RESUMO

Growing tumors exist in metabolically compromised environments that require activation of multiple pathways to scavenge nutrients to support accelerated rates of growth. The folliculin (FLCN) tumor suppressor complex (FLCN, FNIP1, FNIP2) is implicated in the regulation of energy homeostasis via 2 metabolic master kinases: AMPK and mTORC1. Loss-of-function mutations of the FLCN tumor suppressor complex have only been reported in renal tumors in patients with the rare Birt-Hogg-Dube syndrome. Here, we revealed that FLCN, FNIP1, and FNIP2 are downregulated in many human cancers, including poor-prognosis invasive basal-like breast carcinomas where AMPK and TFE3 targets are activated compared with the luminal, less aggressive subtypes. FLCN loss in luminal breast cancer promoted tumor growth through TFE3 activation and subsequent induction of several pathways, including autophagy, lysosomal biogenesis, aerobic glycolysis, and angiogenesis. Strikingly, induction of aerobic glycolysis and angiogenesis in FLCN-deficient cells was dictated by the activation of the PGC-1α/HIF-1α pathway, which we showed to be TFE3 dependent, directly linking TFE3 to Warburg metabolic reprogramming and angiogenesis. Conversely, FLCN overexpression in invasive basal-like breast cancer models attenuated TFE3 nuclear localization, TFE3-dependent transcriptional activity, and tumor growth. These findings support a general role of a deregulated FLCN/TFE3 tumor suppressor pathway in human cancers.


Assuntos
Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos/fisiologia , Neoplasias da Mama/patologia , Neovascularização Patológica/prevenção & controle , Proteínas Proto-Oncogênicas/fisiologia , Proteínas Supressoras de Tumor/fisiologia , Efeito Warburg em Oncologia , Proteínas Quinases Ativadas por AMP/fisiologia , Linhagem Celular Tumoral , Feminino , Humanos , Fosforilação Oxidativa
5.
Nucleic Acids Res ; 49(22): 12706-12715, 2021 12 16.
Artigo em Inglês | MEDLINE | ID: mdl-34791430

RESUMO

Endogenous retroviruses (ERVs) are subject to transcriptional repression in adult tissues, in part to prevent autoimmune responses. However, little is known about the epigenetic silencing of ERV expression. Here, we describe a new role for inhibitor of growth family member 3 (ING3), to add to an emerging group of ERV transcriptional regulators. Our results show that ING3 binds to several ERV promoters (for instance MER21C) and establishes an EZH2-mediated H3K27 trimethylation modification. Loss of ING3 leads to decreases of H3K27 trimethylation enrichment at ERVs, induction of MDA5-MAVS-interferon signaling, and functional inhibition of several virus infections. These data demonstrate an important new function of ING3 in ERV silencing and contributing to innate immune regulation in somatic cells.


Assuntos
Retrovirus Endógenos , Inativação Gênica , Proteínas de Homeodomínio/fisiologia , Imunidade Inata/genética , Proteínas Supressoras de Tumor/fisiologia , Sistemas CRISPR-Cas , Células HT29 , Células HeLa , Código das Histonas , Proteínas de Homeodomínio/metabolismo , Humanos , Proteínas Supressoras de Tumor/metabolismo
6.
Theranostics ; 11(19): 9452-9469, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34646380

RESUMO

Background: Neuronal death is a major hallmark of Alzheimer's disease (AD). Necroptosis, as a programmed necrotic process, is activated in AD. However, what signals and factors initiate necroptosis in AD is largely unknown. Methods: We examined the expression levels of critical molecules in necroptotic signaling pathway by immunohistochemistry (IHC) staining and immunoblotting using brain tissues from AD patients and AD mouse models of APP/PS1 and 5×FAD. We performed brain stereotaxic injection with recombinant TNF-α, anti-TNFR1 neutralizing antibody or AAV-mediated gene expression and knockdown in APP/PS1 mice. For in vitro studies, we used TNF-α combined with zVAD-fmk and Smac mimetic to establish neuronal necroptosis models and utilized pharmacological or molecular biological approaches to study the signaling pathways. Results: We find that activated neuronal necroptosis is dependent on upstream TNF-α/TNFR1 signaling in both neuronal cell cultures and AD mouse models. Upon TNF-α stimulation, accumulated p62 recruits RIPK1 and induces its self-oligomerization, and activates downstream RIPK1/RIPK3/MLKL cascade, leading to neuronal necroptosis. Ectopic accumulation of p62 is caused by impaired autophagy flux, which is mediated by UVRAG downregulation during the TNF-α-promoted necroptosis. Notably, UVRAG overexpression inhibits neuronal necroptosis in cell and mouse models of AD. Conclusions: We identify a finely controlled regulation of neuronal necroptosis in AD by coordinated TNF-α signaling, RIPK1/3 activity and autophagy machinery. Strategies that could fine-tune necroptosis and autophagy may bring in promising therapeutics for AD.


Assuntos
Doença de Alzheimer/fisiopatologia , Fator de Necrose Tumoral alfa/metabolismo , Proteínas Supressoras de Tumor/fisiologia , Doença de Alzheimer/metabolismo , Animais , Apoptose/efeitos dos fármacos , Proteínas Reguladoras de Apoptose/metabolismo , Autofagia/efeitos dos fármacos , Encéfalo/patologia , Morte Celular/efeitos dos fármacos , Modelos Animais de Doenças , Expressão Gênica , Humanos , Camundongos , Necroptose/fisiologia , Necrose/metabolismo , Neurônios/metabolismo , Proteínas de Ligação a RNA/metabolismo , Proteína Serina-Treonina Quinases de Interação com Receptores/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transcriptoma/genética , Proteínas Supressoras de Tumor/metabolismo
7.
FEBS Open Bio ; 11(12): 3230-3236, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34597467

RESUMO

Mitophagy is a form of autophagy specialized to selectively remove mitochondria. Although the PINK1/Parkin pathway is the best described mitophagy of damaged mitochondria, receptor/mediated mitophagy seems to have a pivotal role in cellular development and specialization. The most studied mitophagy receptor BCL2/adenovirus E1B 19-kDa-interacting protein 3-like (BNIP3L/NIX) is shown to be important for the programmed removal of healthy mitochondria during terminal differentiation of erythrocytes, but its role has been proven in various cell types. Despite recent advances in our understanding of its regulation by phosphorylation and dimerization, there remain numerous questions on how BNIP3L/NIX tightly balances between cellular life and death decisions. This brief review intends to summarize ongoing dilemmas related to BNIP3L/NIX.


Assuntos
Proteínas de Membrana/metabolismo , Mitofagia/fisiologia , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Proteínas Reguladoras de Apoptose/metabolismo , Autofagia/fisiologia , Diferenciação Celular , Humanos , Proteínas de Membrana/fisiologia , Mitocôndrias/metabolismo , Proteínas Mitocondriais/metabolismo , Fosforilação , Proteínas Proto-Oncogênicas/fisiologia , Proteínas Supressoras de Tumor/fisiologia , Ubiquitina-Proteína Ligases
8.
Front Endocrinol (Lausanne) ; 12: 700083, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34552555

RESUMO

Primary cilia are sensory organelles with a variety of receptors and channels on their membranes. Recently, primary cilia were proposed to be crucial sites for exocytosis and endocytosis of vesicles associated with endocytic control of various ciliary signaling pathways. Thyroglobulin (Tg) synthesis and Tg exocytosis/endocytosis are critical for the functions of thyroid follicular cells, where primary cilia are relatively well preserved. LRP2/megalin has been detected on the apical surface of absorptive epithelial cells, including thyrocytes. LRP2/megalin on thyrocytes serves as a Tg receptor and can mediate Tg endocytosis. In this study, we investigated the role of primary cilia in LRP2/megalin expression in thyroid gland stimulated with endogenous TSH using MMI-treated and Tg-Cre;Ift88flox/flox mice. LRP2/megalin expression in thyroid follicles was higher in MMI-treated mice than in untreated control mice. MMI-treated mice exhibited a significant increase in ciliogenesis in thyroid follicular cells relative to untreated controls. Furthermore, MMI-induced ciliogenesis accompanied increases in LRP2/megalin expression in thyroid follicular cells, in which LRP2/megalin was localized to the primary cilium. By contrast, in Tg-Cre;Ift88flox/flox mice, thyroid with defective primary cilia expressed markedly lower levels of LRP2/megalin. Serum Tg levels were elevated in MMI-treated mice and reduced in Tg-Cre;Ift88flox/flox mice. Taken together, these results indicate that defective ciliogenesis in murine thyroid follicular cells is associated with impaired LRP2/megalin expression and reduced serum Tg levels. Our results strongly suggest that primary cilia harbors LRP2/megalin, and are involved in TSH-mediated endocytosis of Tg in murine thyroid follicles.


Assuntos
Cílios/fisiologia , Endocitose , Proteína-2 Relacionada a Receptor de Lipoproteína de Baixa Densidade/metabolismo , Tireoglobulina/metabolismo , Glândula Tireoide/metabolismo , Tireotropina/farmacologia , Proteínas Supressoras de Tumor/fisiologia , Animais , Proteína-2 Relacionada a Receptor de Lipoproteína de Baixa Densidade/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptores de Superfície Celular/genética , Receptores de Superfície Celular/metabolismo , Glândula Tireoide/efeitos dos fármacos
9.
Med Oncol ; 38(11): 132, 2021 Sep 28.
Artigo em Inglês | MEDLINE | ID: mdl-34581882

RESUMO

WAC is closely related to the occurrence and development of tumors. However, its role in human glioblastoma (GBM) and its potential regulatory mechanisms have not been investigated. This study demonstrated that WAC is downregulated in GBM, and its low expression predicts a poor prognosis. We investigated the effect of WAC on the proliferation of glioma cells through a CCK-8 assay, EdU incorporation, and cell formation. The effects of WAC on apoptosis and autophagy in glioma were determined by flow cytometry, TUNEL detection, immunofluorescence, q-PCR, WB, and scanning electron microscopy. We found that overexpression of WAC inhibited the proliferation of glioma cells, promoted apoptosis, and induced autophagy. Therefore, WAC is likely to play a role as a new regulatory molecule in glioma.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/fisiologia , Apoptose , Autofagia/fisiologia , Neoplasias Encefálicas/patologia , Glioblastoma/patologia , Proteínas Supressoras de Tumor/fisiologia , Neoplasias Encefálicas/mortalidade , Neoplasias Encefálicas/prevenção & controle , Linhagem Celular Tumoral , Proliferação de Células , Glioblastoma/mortalidade , Glioblastoma/prevenção & controle , Humanos , Transdução de Sinais
10.
J BUON ; 26(4): 1498-1503, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34565010

RESUMO

PURPOSE: This study aimed to detect the differential expression of microRNA-363-3p (miR-363-3p) in bladder cancer (BC) samples and to explore its influence on metastasis of BC cells. METHODS: Expression level of miR-363-3p in 70 cases of BC tissues and paracancerous tissues was detected. After establishing miR-363-3p overexpression model in 253j and RT4 cells, their migratory ability was assessed by Transwell and wound healing assay. The interaction between miR-363-3p and its downstream gene was predicted online and further confirmed by luciferase assay. Their involvement in regulating metastasis of BC cells was finally explored. RESULTS: MiR-363-3p was downregulated in BC tissues compared with that in the paracancerous tissues. Overexpression of miR-363-3p markedly weakened migratory ability in BC cells. BTG2 was the downstream gene binding miR-363-3p. In addition, overexpression of BTG2 reversed the inhibitory effect of miR-363-3p on BC cell migration. CONCLUSIONS: MiR-363-3p is lowly expressed in BC samples. It weakens in vitro migratory ability in BC cells through downregulating BTG2.


Assuntos
Proteínas Imediatamente Precoces/fisiologia , MicroRNAs/fisiologia , Proteínas Supressoras de Tumor/fisiologia , Neoplasias da Bexiga Urinária/genética , Neoplasias da Bexiga Urinária/patologia , Movimento Celular/genética , Regulação para Baixo , Regulação Neoplásica da Expressão Gênica , Humanos , Metástase Neoplásica/genética , Células Tumorais Cultivadas
11.
Mol Biol Cell ; 32(21): ar23, 2021 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-34495684

RESUMO

Scribble (Scrib), Discs-large (Dlg), and Lethal giant larvae (Lgl) are basolateral regulators of epithelial polarity and tumor suppressors whose molecular mechanisms of action remain unclear. We used proximity biotinylation to identify proteins localized near Dlg in the Drosophila wing imaginal disc epithelium. In addition to expected membrane- and cytoskeleton-associated protein classes, nuclear proteins were prevalent in the resulting mass spectrometry dataset, including all four members of the nucleosome remodeling factor (NURF) chromatin remodeling complex. Subcellular fractionation demonstrated a nuclear pool of Dlg and proximity ligation confirmed its position near the NURF complex. Genetic analysis showed that NURF activity is also required for the overgrowth of dlg tumors, and this growth suppression correlated with a reduction in Hippo pathway gene expression. Together, these data suggest a nuclear role for Dlg in regulating chromatin and transcription through a more direct mechanism than previously thought.


Assuntos
Proteínas de Drosophila/metabolismo , Complexo Mi-2 de Remodelação de Nucleossomo e Desacetilase/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Animais , Polaridade Celular/genética , Proteínas do Citoesqueleto/metabolismo , Proteínas de Drosophila/fisiologia , Drosophila melanogaster , Células Epiteliais/metabolismo , Epitélio , Discos Imaginais/metabolismo , Proteínas de Membrana/metabolismo , Complexo Mi-2 de Remodelação de Nucleossomo e Desacetilase/genética , Proteínas Nucleares/metabolismo , Proteína Quinase C/metabolismo , Fatores de Transcrição/metabolismo , Proteínas Supressoras de Tumor/fisiologia
12.
Kaohsiung J Med Sci ; 37(8): 675-685, 2021 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-34337857

RESUMO

Lung adenocarcinoma (LUAD) is the most common type of lung cancer. Currently, the survival rate of LUAD patients remains low due to heterogeneity and high invasiveness. The long non-coding RNA (lncRNA) cancer susceptibility candidate 2 (CASC2) is reported to be related to LUAD development. Hence, we investigate the roles and regulatory mechanism of CASC2 in LUAD. The expression levels of CASC2, microRNA (miR)-21, and p53 were quantified by quantitative real-time polymerase chain reaction, and the protein levels of Bax, Bcl-2, p53, and p21 were examined using western blotting. A dual-luciferase reporter experiment was conducted to prove the molecular interactions between CASC2 and miR-21 or p53. CCK-8 and flow cytometry assays were conducted to assess cell proliferation and apoptosis, respectively. CASC2 was expressed at a low level in LUAD patients and LUAD cell lines. CASC2 overexpression markedly suppressed cell proliferation and enhanced apoptosis. Mechanistically, CASC2 overexpression dramatically inhibited miR-21 expression and increased p53 expression by directly targeting miR-21. Moreover, rescue experiments suggested that either miR-21 overexpression or p53 silencing obviously weakened the biological effects of CASC2 overexpression. In addition, p53 was proven to be an upstream transcription factor of CASC2 and can activate CASC2 transcription. These results provide evidence that the lncRNA CASC2/miR-21/p53 form a positive feedback loop to mediate cell proliferation and apoptosis in LUAD, which may provide a new insight into the pathological mechanisms of LUAD.


Assuntos
Adenocarcinoma de Pulmão/patologia , Neoplasias Pulmonares/patologia , MicroRNAs/fisiologia , Proteína Supressora de Tumor p53/fisiologia , Proteínas Supressoras de Tumor/fisiologia , Adenocarcinoma de Pulmão/genética , Progressão da Doença , Humanos , Neoplasias Pulmonares/genética
13.
PLoS Genet ; 17(8): e1009753, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-34388164

RESUMO

Meiosis is essential for the generation of gametes and sexual reproduction, yet the factors and underlying mechanisms regulating meiotic progression remain largely unknown. Here, we showed that MTL5 translocates into nuclei of spermatocytes during zygotene-pachytene transition and ensures meiosis advances beyond pachytene stage. MTL5 shows strong interactions with MuvB core complex components, a well-known transcriptional complex regulating mitotic progression, and the zygotene-pachytene transition of MTL5 is mediated by its direct interaction with the component LIN9, through MTL5 C-terminal 443-475 residues. Male Mtl5c-mu/c-mu mice expressing the truncated MTL5 (p.Ser445Arg fs*3) that lacks the interaction with LIN9 and is detained in cytoplasm showed male infertility and spermatogenic arrest at pachytene stage, same as that of Mtl5 knockout mice, indicating that the interaction with LIN9 is essential for the nuclear translocation and function of MTL5 during meiosis. Our data demonstrated MTL5 translocates into nuclei during the zygotene-pachytene transition to initiate its function along with the MuvB core complex in pachytene spermatocytes, highlighting a new mechanism regulating the progression of male meiosis.


Assuntos
Meiose/fisiologia , Metalotioneína/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Transporte Ativo do Núcleo Celular/fisiologia , Animais , Proteínas de Ciclo Celular/metabolismo , Pareamento Cromossômico/genética , Citoplasma , Proteínas de Ligação a DNA , Fertilidade/genética , Fertilidade/fisiologia , Infertilidade Masculina/genética , Infertilidade Masculina/metabolismo , Masculino , Prófase Meiótica I/fisiologia , Metalotioneína/genética , Camundongos , Camundongos Endogâmicos C57BL , Estágio Paquíteno/genética , Espermatócitos/fisiologia , Espermatogênese/fisiologia , Testículo , Proteínas Supressoras de Tumor/fisiologia
14.
Cancer Sci ; 112(10): 4064-4074, 2021 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-34251713

RESUMO

Programmed cell death ligand 1 (PD-L1) is a major immunosuppressive checkpoint protein expressed by tumor cells to subvert anticancer immunity. Recent studies have shown that ionizing radiation (IR) upregulates the expression of PD-L1 in tumor cells. However, whether an IR-induced DNA damage response (DDR) directly regulates PD-L1 expression and the functional significance of its upregulation are not fully understood. Here, we show that IR-induced upregulation of PD-L1 expression proceeds through both transcriptional and post-translational mechanisms. Upregulated PD-L1 was predominantly present on the cell membrane, resulting in T-cell apoptosis in a co-culture system. Using mass spectrometry, we identified PD-L1 interacting proteins and found that BCLAF1 (Bcl2 associated transcription factor 1) is an important regulator of PD-L1 in response to IR. BCLAF1 depletion decreased PD-L1 expression by promoting the ubiquitination of PD-L1. In addition, we show that CMTM6 is upregulated in response to IR and participates in BCLAF1-dependent PD-L1 upregulation. Finally, we demonstrated that the ATM/BCLAF1/PD-L1 axis regulated PD-L1 stabilization in response to IR. Together, our findings reveal a novel regulatory mechanism of PD-L1 expression in the DDR.


Assuntos
Antígeno B7-H1/metabolismo , Radiação Ionizante , Proteínas Repressoras/fisiologia , Proteínas Supressoras de Tumor/fisiologia , Apoptose , Proteínas Mutadas de Ataxia Telangiectasia/metabolismo , Antígeno B7-H1/efeitos da radiação , Linhagem Celular Tumoral , Membrana Celular/metabolismo , Técnicas de Cocultura , Dano ao DNA , Humanos , Células Jurkat , Proteínas com Domínio MARVEL/metabolismo , Proteínas com Domínio MARVEL/efeitos da radiação , Espectrometria de Massas , Proteínas da Mielina/metabolismo , Proteínas da Mielina/efeitos da radiação , Proteínas de Neoplasias/metabolismo , Modificação Traducional de Proteínas , Processamento de Proteína Pós-Traducional , Proteínas Repressoras/deficiência , Linfócitos T/citologia , Linfócitos T/efeitos da radiação , Proteínas Supressoras de Tumor/deficiência , Ubiquitinação , Regulação para Cima/efeitos da radiação
15.
Biosystems ; 208: 104475, 2021 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-34237349

RESUMO

The Hippo signaling primarily includes LATS1/2 and YAP1. Recent work has demonstrated a novel negative feedback between YAP1 and LATS1/2. However, how YAP-LATS negative feedback regulates cancer progression remains elusive. We constructed a multi-scale model which integrates angiogenesis, spatiotemporal variation of microenvironmental factors and phenotypic switch of tumor cells. Our simulation replicated the findings that YAP overexpression markedly attenuated cell proliferation owing to elevated negative feedback strength. After disruption of YAP-LATS negative feedback loop, however, YAP overexpression would promote cell proliferation. Consistently, LATS overexpression inhibited cell growth and lowered the proliferation potential. We also employed a putative LATS agonist and identified its dose-dependent tumor suppressive effects. Furthermore, targeted delivery could more effectively inhibit tumor growth. Our model has reconciled experimental findings and implied that reconstruction of functional and/or hyperactivated YAP-LATS negative feedback might be a promising strategy to homeostatic maintenance and tumor suppression.


Assuntos
Retroalimentação Fisiológica/fisiologia , Via de Sinalização Hippo/fisiologia , Homeostase/fisiologia , Modelos Biológicos , Proteínas Supressoras de Tumor/fisiologia , Proteínas de Sinalização YAP/fisiologia , Animais , Proliferação de Células/fisiologia , Humanos , Neoplasias/patologia , Proteínas Serina-Treonina Quinases/fisiologia
16.
Mol Cell ; 81(17): 3526-3541.e8, 2021 09 02.
Artigo em Inglês | MEDLINE | ID: mdl-34186021

RESUMO

BAP1 is mutated or deleted in many cancer types, including mesothelioma, uveal melanoma, and cholangiocarcinoma. It is the catalytic subunit of the PR-DUB complex, which removes PRC1-mediated H2AK119ub1, essential for maintaining transcriptional repression. However, the precise relationship between BAP1 and Polycombs remains elusive. Using embryonic stem cells, we show that BAP1 restricts H2AK119ub1 deposition to Polycomb target sites. This increases the stability of Polycomb with their targets and prevents diffuse accumulation of H2AK119ub1 and H3K27me3. Loss of BAP1 results in a broad increase in H2AK119ub1 levels that is primarily dependent on PCGF3/5-PRC1 complexes. This titrates PRC2 away from its targets and stimulates H3K27me3 accumulation across the genome, leading to a general chromatin compaction. This provides evidence for a unifying model that resolves the apparent contradiction between BAP1 catalytic activity and its role in vivo, uncovering molecular vulnerabilities that could be useful for BAP1-related pathologies.


Assuntos
Cromatina/metabolismo , Proteínas do Grupo Polycomb/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Ubiquitina Tiolesterase/metabolismo , Animais , Linhagem Celular/metabolismo , Cromatina/genética , Cromatina/fisiologia , Células-Tronco Embrionárias/metabolismo , Heterocromatina , Histonas/metabolismo , Humanos , Camundongos , Células-Tronco Embrionárias Murinas/metabolismo , Complexo Repressor Polycomb 1/metabolismo , Complexo Repressor Polycomb 2/metabolismo , Proteínas do Grupo Polycomb/genética , Proteínas Supressoras de Tumor/genética , Proteínas Supressoras de Tumor/fisiologia , Ubiquitina Tiolesterase/genética , Ubiquitina Tiolesterase/fisiologia , Ubiquitinação
17.
J Invest Dermatol ; 141(10): 2354-2368, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-33845078

RESUMO

Cutaneous squamous cell carcinomas (SCCs) are frequent heterogeneous tumors arising from sun-exposed regions of the skin and characterized by complex pathogenesis. HOPX is a member of the homeodomain-containing superfamily of proteins holding an atypical homeodomain unable to bind to DNA. First discovered in the heart as a regulator of cardiac development, in the skin, HOPX modulates the terminal differentiation of keratinocytes. There is a particular interest in studying HOPX in squamous skin carcinogenesis because it has the atypical structure and the functional duality as an oncogene and a tumor suppressor gene, reported in different malignancies. In this study, we analyzed the effects of HOPX knockdown and overexpression on SCC tumorigenicity in vitro and in vivo. Our data show that HOPX knockdown in SCC cells inhibits their proliferative and invasive activity through the acceleration of apoptosis. We established that methylation of two alternative HOPX promoters leads to differential expression of HOPX transcripts in normal keratinocytes and SCC cells. Importantly, we report that HOPX acts as an oncogene in the pathogenesis of SCC probably through the activation of the second alternative promoter and the modulation of apoptosis.


Assuntos
Carcinoma de Células Escamosas/etiologia , Proteínas de Homeodomínio/fisiologia , Neoplasias Cutâneas/etiologia , Proteínas Supressoras de Tumor/fisiologia , Animais , Apoptose , Carcinogênese , Carcinoma de Células Escamosas/patologia , Proliferação de Células , Células Cultivadas , Metilação de DNA , Feminino , Proteínas de Homeodomínio/genética , Humanos , Camundongos , Oncogenes , Regiões Promotoras Genéticas , Neoplasias Cutâneas/patologia , Sítio de Iniciação de Transcrição , Proteínas Supressoras de Tumor/genética
18.
J Invest Dermatol ; 141(10): 2436-2448, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-33864770

RESUMO

Many inflammatory skin diseases are characterized by altered epidermal differentiation. Whether this altered differentiation promotes inflammatory responses has been unknown. Here, we show that IRAK2, a member of the signaling complex downstream of IL-1 and IL-36, correlates positively with disease severity in both atopic dermatitis and psoriasis. Inhibition of epidermal IRAK2 normalizes differentiation and inflammation in two mouse models of psoriasis- and atopic dermatitis-like inflammation. Specifically, we demonstrate that IRAK2 ties together proinflammatory and differentiation-dependent responses and show that this function of IRAK2 is specific to keratinocytes and acts through the differentiation-associated transcription factor ZNF750. Taken together, our findings suggest that IRAK2 has a critical role in promoting feed-forward amplification of inflammatory responses in skin through modulation of differentiation pathways and inflammatory responses.


Assuntos
Epiderme/patologia , Inflamação/etiologia , Quinases Associadas a Receptores de Interleucina-1/fisiologia , Diferenciação Celular , Células Cultivadas , Dermatite Atópica/etiologia , Humanos , NF-kappa B/fisiologia , Psoríase/etiologia , Índice de Gravidade de Doença , Transdução de Sinais , Fatores de Transcrição/fisiologia , Proteínas Supressoras de Tumor/fisiologia
19.
Cell Tissue Res ; 384(3): 643-653, 2021 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-33783611

RESUMO

Individual olfactory sensory neurons (OSNs) in the mouse main olfactory epithelium express a single odorant receptor (OR) gene from the repertoire of either class I or class II ORs. The transcription factor Bcl11b determines the OR class to be expressed in OSNs. The septal organ (SO), a small neuroepithelium located at the ventral base of the nasal septum, is considered as an olfactory subsystem because it expresses a specific subset of ORs. However, the mechanisms underlying the generation and differentiation of SO-OSN remain unknown. In the present study, we show that the generation and differentiation of SO-OSN employ the same genetic pathway as in the OSN lineage, which is initiated by the neuronal fate determinant factor Ascl1. Additionally, the key role of Bcl11b in the SO is demonstrated by the abnormal phenotypes of Bcl11b-deficient mice: significant reduction in the expression of OR genes and in the number of mature SO-OSNs. Although SO-OSNs are specified to express a subset of class II OR genes in wild-type mice, the Bcl11b deletion led to the expression of class I OR genes, while the expression of class II OR genes was significantly decreased, with one exception of Olfr15. These results indicate that Bcl11b is necessary for proper OR expression in SO-OSNs.


Assuntos
Perfuração do Septo Nasal/metabolismo , Mucosa Olfatória/metabolismo , Neurônios Receptores Olfatórios/metabolismo , Receptores Odorantes/metabolismo , Proteínas Repressoras/fisiologia , Proteínas Supressoras de Tumor/fisiologia , Animais , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mucosa Olfatória/citologia , Neurônios Receptores Olfatórios/citologia , Olfato
20.
Cancer Lett ; 506: 67-82, 2021 05 28.
Artigo em Inglês | MEDLINE | ID: mdl-33675985

RESUMO

Very limited progress has been made in the management of advanced melanoma, especially melanoma of uveal origin. Lactamase ß (LACTB) is a novel tumor suppressor; however, its biological function in melanoma remains unknown. Herein we demonstrated markedly lower LACTB expression levels in melanoma tissues and cell lines. Overexpression of LACTB suppressed the proliferation, migration and invasion of melanoma cells in vitro. Mechanistically, LACTB inhibited the activity of yes-associated protein (YAP). We showed that the level of phospho-YAP (Serine 127) was increased upon LACTB overexpression, which prevented the translocation of YAP to the nucleus. Further, LACTB could directly bind to PP1A and attenuate the interaction between PP1A and YAP, resulting in decreased YAP dephosphorylation and inactivation in a LATS1-independent manner. Additionally, transfection of phosphorylation-defective YAP mutants reversed LACTB-induced tumor suppression. Upstream, we demonstrated that SOX10 binds to the LACTB promoter and negatively regulates its transcription. Overexpression of LACTB also suppressed the tumorigenicity and lung metastasis of MUM2B uveal melanoma cells in vivo. Taken together, our findings indicate a novel SOX10/LACTB/PP1A signaling cascade that renders YAP inactive and modulates melanoma progression, offering a new therapeutic target for melanoma treatment.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/fisiologia , Melanoma/prevenção & controle , Proteínas de Membrana/fisiologia , Proteínas Mitocondriais/fisiologia , Proteína Fosfatase 1/fisiologia , Fatores de Transcrição/fisiologia , Proteínas Supressoras de Tumor/fisiologia , beta-Lactamases/fisiologia , Animais , Linhagem Celular Tumoral , Humanos , Neoplasias Pulmonares/secundário , Melanoma/patologia , Proteínas de Membrana/genética , Camundongos , Proteínas Mitocondriais/genética , Fosforilação , Proteínas Serina-Treonina Quinases/fisiologia , Fatores de Transcrição SOXE/fisiologia , Ensaios Antitumorais Modelo de Xenoenxerto , Proteínas de Sinalização YAP , beta-Lactamases/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...