Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
J Virol ; : e0095124, 2024 Sep 17.
Artigo em Inglês | MEDLINE | ID: mdl-39287390

RESUMO

The mRNAs encoding the human papillomavirus type 16 (HPV16) E6 and E7 oncogene mRNAs are subjected to extensive alternative RNA splicing at multiple regulated splice sites. One of the most extensively used 5'-splice sites in the HPV16 genome is named SD880 and is located immediately downstream of the E7 open reading frame. Here, we show that a cluster of three GGG-motifs adjacent to HPV16 SD880 interacts with heterogeneous nuclear ribonucleoprotein (hnRNP) H that cooperates with SD880 to stimulate splicing to the upstream HPV16 3'-splice site SA742. This splice site is located in the E7 coding region and is required for the production of the HPV16 226^742 mRNA that encodes the E6^E7 fusion protein. Enhancement of HPV16 E6^E7 mRNA production by hnRNP H occurred at the expense of the intron-retained E6 mRNAs and the spliced E7 mRNAs, demonstrating that hnRNP H controls the relative levels of E6, E7, and E6^E7 proteins. Unexpectedly, overexpression of hnRNP H also promoted retention of the downstream E1 encoding intron and enhanced E1 protein production. We concluded that hnRNP H plays an important role in the HPV16 gene expression program.IMPORTANCEHere, we show that hnRNP H binds to multiple GGG-motifs downstream of human papillomavirus type 16 (HPV16) splice site SD880 and acts in concert with SD880 to promote expression of the HPV16 E6^E7 mRNA. The E6^E7 protein has been shown previously to stabilize the HPV16 E6 and E7 oncoproteins and may as such contribute to the carcinogenic properties of HPV16. In its capacity of major regulator of HPV16 oncogene expression, hnRNP H may be exploited as a target for antiviral drugs to HPV16.

2.
Pharmacol Res ; 207: 107341, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-39134188

RESUMO

Drug conjugates are obtained from tumor-located vectors connected to cytotoxic agents via linkers, which are designed to deliver hyper-toxic payloads directly to targeted cancer cells. These drug conjugates include antibody-drug conjugates (ADCs), peptide-drug conjugates (PDCs), small molecule-drug conjugates (SMDCs), nucleic acid aptamer-drug conjugates (ApDCs), and virus-like drug conjugate (VDCs), which show great therapeutic value in the clinic. Drug conjugates consist of a targeting carrier, a linker, and a payload. Payloads are key therapy components. Cytotoxic molecules and their derivatives derived from natural products are commonly used in the payload portion of conjugates. The ideal payload should have sufficient toxicity, stability, coupling sites, and the ability to be released under specific conditions to kill tumor cells. Microtubule protein inhibitors, DNA damage agents, and RNA inhibitors are common cytotoxic molecules. Among these conjugates, cytotoxic molecules of natural origin are summarized based on their mechanism of action, conformational relationships, and the discovery of new derivatives. This paper also mentions some cytotoxic molecules that have the potential to be payloads. It also summarizes the latest technologies and novel conjugates developed in recent years to overcome the shortcomings of ADCs, PDCs, SMDCs, ApDCs, and VDCs. In addition, this paper summarizes the clinical trials conducted on conjugates of these cytotoxic molecules over the last five years. It provides a reference for designing and developing safer and more efficient conjugates.


Assuntos
Antineoplásicos , Produtos Biológicos , Imunoconjugados , Neoplasias , Humanos , Neoplasias/tratamento farmacológico , Animais , Produtos Biológicos/uso terapêutico , Produtos Biológicos/química , Produtos Biológicos/farmacologia , Antineoplásicos/uso terapêutico , Antineoplásicos/química , Antineoplásicos/farmacologia , Imunoconjugados/uso terapêutico , Imunoconjugados/química , Imunoconjugados/farmacologia
4.
Biomed Res Int ; 2021: 5596014, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34055985

RESUMO

Osteosarcoma is the most common type of bone malignancy, and the pathogenesis has not been entirely elucidated yet. An important deimination modification enzyme PADI4 (peptidylarginine deiminase 4) has attracted much attention in recent years for its important function in several kinds of human tumors. However, the role of PADI4 on osteosarcoma tumorigenesis remains largely unrevealed. Here, we first assessed the effect of PADI4 on osteosarcoma proliferation by the CCK8 method and colony formation assay. Ectopically expressing PADI4 positively regulates the colony formation capacity of both U2OS and Saos-2 cells. Furthermore, we explored the related mechanism and showed that PADI4 could stimulate Wnt/ß-catenin and MEK/ERK signaling in both U2OS and Saos-2 cells. Then, we detected expression of PADI4 in human tissues of osteosarcoma and revealed that differential expression of PADI4 was associated with tumorigenesis of osteosarcoma. Last, we performed the in vivo experiment in nude mice and results also showed PADI4 could affect the tumor growth. In conclusion, this work revealed that PADI4 could upregulate the proliferation of osteosarcoma, mainly via the Wnt/ß-catenin and MEK/ERK signaling pathway. This study gives us new insight into the regulation mechanism of osteosarcoma proliferation and highlights PADI4 as a promising target for osteosarcoma diagnosis and treatment.


Assuntos
Neoplasias Ósseas/metabolismo , Proliferação de Células/efeitos dos fármacos , Osteossarcoma/metabolismo , Proteína-Arginina Desiminase do Tipo 4/metabolismo , Proteína-Arginina Desiminase do Tipo 4/farmacologia , Adolescente , Adulto , Idoso , Animais , Neoplasias Ósseas/patologia , Carcinogênese , Linhagem Celular Tumoral , Colecistocinina , Feminino , Humanos , Masculino , Camundongos , Camundongos Nus , Pessoa de Meia-Idade , Osteossarcoma/patologia , Fragmentos de Peptídeos , Proteína-Arginina Desiminase do Tipo 4/genética , Desiminases de Arginina em Proteínas , Transdução de Sinais , Ensaios Antitumorais Modelo de Xenoenxerto , Adulto Jovem , beta Catenina/metabolismo
5.
J Cell Mol Med ; 24(16): 9135-9144, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32588529

RESUMO

Recent studies have shown that tumour necrosis factor-α-induced protein 8 like-1(TIPE1) plays distinct roles in different cancers. TIPE1 inhibits tumour proliferation and metastasis in a variety of tumours but acts as an oncogene in cervical cancer. The role of TIPE1 in nasopharyngeal carcinoma (NPC) remains unknown. Interestingly, TIPE1 expression was remarkably increased in NPC tissue samples compared to adjacent normal nasopharyngeal epithelial tissue samples in our study. TIPE1 expression was positively correlated with that of the proliferation marker Ki67 and negatively correlated with patient lifespan. In vitro, TIPE1 inhibited autophagy and induced cell proliferation in TIPE1-overexpressing CNE-1 and CNE-2Z cells. In addition, knocking down TIPE1 expression promoted autophagy and decreased proliferation, whereas overexpressing TIPE1 increased the levels of pmTOR, pS6 and P62 and decreased the level of pAMPK and the LC3B. Furthermore, the decrease in autophagy was remarkably rescued in TIPE1-overexpressing CNE-1 and CNE-2Z cells treated with the AMPK activator AICAR. In addition, TIPE1 promoted tumour growth in BALB/c nude mice. Taken together, results indicate that TIPE1 promotes NPC progression by inhibiting autophagy and inducing cell proliferation via the AMPK/mTOR signalling pathway. Thus, TIPE1 could potentially be used as a valuable diagnostic and prognostic biomarker for NPC.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Autofagia/fisiologia , Proliferação de Células/fisiologia , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Carcinoma Nasofaríngeo/metabolismo , Neoplasias Nasofaríngeas/metabolismo , Transdução de Sinais/fisiologia , Serina-Treonina Quinases TOR/metabolismo , Animais , Biomarcadores Tumorais/metabolismo , Linhagem Celular Tumoral , Humanos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Ensaios Antitumorais Modelo de Xenoenxerto/métodos
6.
Life Sci ; 256: 117968, 2020 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-32544462

RESUMO

Osteosarcoma (OS) is the most common type of primary bone malignancy with high recurrence and metastasis. Peptidylarginine deiminase 4 (PADI4), as an important protein post-translational modification enzyme, has been identified as a potential regulator in the invasion and migration in several types of tumors. The role of PADI4 in osteosarcoma metastasis remains unknown. In this study, we revealed significant positive correlation between PADI4 and pulmonary metastasis of osteosarcoma. Wound-healing and transwell assay indicated that PADI4 induced invasion and migration of osteosarcoma cell in vitro while PADI4 inhibitor has repressive effect. PADI4 mutation with no deimination activity exhibited no significant effect on invasion and migration of osteosarcoma cells. Moreover, we evaluated the effect of PADI4 on expression of the markers of epithelial-mesenchymal transition and results showed that PADI4 promoted EMT while PADI4 inhibitor suppressed EMT in osteosarcoma cells. We also detected the expression of PADI4 and E-Cadherin in the tissues of osteosarcoma patients with or without pulmonary metastasis. Results showed positive relationship between the expression of PADI4 and osteosarcoma metastasis. In contrast, the expression of E-Cadherin exhibited negative correlation with PADI4 and osteosarcoma metastasis. Our research offered a novel link between PADI4 and osteosarcoma metastasis and demonstrated PADI4 as a promising target for treatment of osteosarcoma metastasis.


Assuntos
Movimento Celular , Regulação para Baixo , Transição Epitelial-Mesenquimal , Osteossarcoma/enzimologia , Osteossarcoma/patologia , Proteína-Arginina Desiminase do Tipo 4/metabolismo , Antígenos CD/metabolismo , Caderinas/metabolismo , Linhagem Celular Tumoral , Regulação para Baixo/genética , Transição Epitelial-Mesenquimal/genética , Regulação Neoplásica da Expressão Gênica , Humanos , Neoplasias Pulmonares/secundário , Invasividade Neoplásica , Osteossarcoma/genética , Proteína-Arginina Desiminase do Tipo 4/genética
7.
Carcinogenesis ; 40(4): 592-599, 2019 06 10.
Artigo em Inglês | MEDLINE | ID: mdl-30445600

RESUMO

Previous studies have shown that TIPE1 inhibits tumor proliferation and metastasis in certain cancers; however, increased expression of TIPE1 is observed in cervical cancer cell lines and tissues, indicating it might exert a distinctive role in cervical cancer. Cell and xenograft tumorigenicity assays showed that TIPE1 facilitates cervical cancer progression in this study. Further investigation demonstrated that TIPE1 binds to p53 and impairs its activity via inhibition of its acetylation. In addition, TIPE1 promoted cell proliferation and suppressed cisplatin susceptibility in a p53-dependent manner, indicating that TIPE1 facilitates cervical cancer progression primarily through the p53 pathway. TIPE1 expression in clinical samples also demonstrated that its upregulation predicts poor prognosis in patients with cervical cancer. Taken together, the results of this study showed that TIPE1 serves as an oncogene by restricting p53 activity in the development of cervical cancer, suggesting that TIPE1 will provide a new potential target for cervical cancer therapy and can be used as a biomarker to predict patient prognosis.


Assuntos
Biomarcadores Tumorais/metabolismo , Regulação Neoplásica da Expressão Gênica , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Neoplasias do Colo do Útero/mortalidade , Neoplasias do Colo do Útero/patologia , Acetilação , Animais , Apoptose , Biomarcadores Tumorais/genética , Proliferação de Células , Progressão da Doença , Feminino , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/genética , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Prognóstico , Taxa de Sobrevida , Células Tumorais Cultivadas , Proteína Supressora de Tumor p53/genética , Neoplasias do Colo do Útero/genética , Neoplasias do Colo do Útero/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
8.
Prostate ; 78(16): 1248-1261, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30105831

RESUMO

BACKGROUND: Prostate cancer (PCa) is a leading cause of death in males all over the world; besides, the diagnosis and therapy of it are still challenging. Researchers have revealed that long non-coding RNAs (lncRNAs) play important roles in the genesis and progression of human cancers, including PCa. METHODS: Bioinformatics analysis and Kaplan-Meier survival analysis were utilized to confirm TMPO-AS1 as a diagnostic and prognostic marker. The TMPO-AS1 levels in both patient tissues and PCa cell lines were determined by qRT-PCR analysis. Moreover, the chromatin immunoprecipitation (ChIP) assay identified that TMPO-AS1 was a direct target of AR. The effect of overexpression or knockdown of TMPO-AS1 on cell proliferation, migration, cell cycle, and cell apoptosis was assessed by using CCK-8, transwell assays, and flow cytometric analysis, respectively. RESULTS: Based on primary screening, we found that TMPO-AS1 could be a useful diagnostic and prognostic marker for PCa, whose expression was upregulated in PCa samples and associated with poorer prognosis. Bioinformatics predictions revealed TMPO-AS1 was associated with a series of biological processes involved in PCa progression. In PCa cells, TMPO-AS1 was predominantly localized in the cytoplasm and directly down-regulated by AR. Gain/loss-of-function assays showed TMPO-AS1 overexpression increased cell proliferation by promoting cell cycle progression and promoted migration, but reduced apoptosis of PCa cells. In addition, TMPO-AS1 may be a diagnostic and prognostic marker in multiple cancer types. CONCLUSIONS: AR-regulated lncRNA TMPO-AS1 functioned as an oncogenic lncRNA in PCa, and may be a potential diagnostic and prognostic biomarker to be used as a therapeutic target for PCa.


Assuntos
Regulação Neoplásica da Expressão Gênica , Neoplasias da Próstata/genética , RNA Longo não Codificante/genética , Apoptose/genética , Ciclo Celular/genética , Linhagem Celular Tumoral , Movimento Celular/genética , Proliferação de Células/genética , Progressão da Doença , Humanos , Masculino , Pessoa de Meia-Idade , Prognóstico , Neoplasias da Próstata/patologia
9.
J Diabetes Res ; 2017: 4802319, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28626770

RESUMO

BACKGROUND: Tumor necrosis factor-α-induced protein 8-like 2 (TIPE2 or TNFAIP8L2) is a negative regulator of natural and adaptive immunity. The role of TIPE2 in type 2 diabetes mellitus (T2DM) remains unknown, although TIPE2 plays key roles in preserving inflammatory homeostasis. METHODS: TIPE2 expression was measured by Western blotting and real-time polymerase chain reaction (RT-PCR) in peripheral blood mononuclear cells (PBMCs) isolated from T2DM patients and healthy controls, and tumor necrosis factor-α (TNF-α), high-sensitivity C-reactive protein (hsCRP), interleukin 6 (IL-6), and other related biometabolic parameters were detected using a nephelometer or by ELISA. Differentiated THP-1 cells were exposed to siTIPE2 and TIPE2 adenovirus. RESULTS: TIPE2 was significantly increased in PBMCs from T2DM patients compared with those from healthy controls and was negatively correlated with serum TNF-α, IL-6, and hsCRP concentrations but positively correlated with HbA1c and LDL-C in T2DM patients. High glucose treatment (50 mmol/L) can upregulate the expression of TIPE2 and cytokine secretion in differentiated THP-1 cells. siTIPE2 infection exacerbated the increased TNF-α and IL-6 concentrations in differentiated THP-1 cells under high glucose conditions (50 mmol/L), while infection with TIPE2 adenovirus reversed the increased TNF-α concentration. CONCLUSIONS: The present study indicates that TIPE2 may participate in T2DM by regulating TNF-α production.


Assuntos
Diabetes Mellitus Tipo 2/sangue , Interleucina-6/sangue , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Fator de Necrose Tumoral alfa/sangue , Imunidade Adaptativa , Idoso , Glicemia/metabolismo , Proteína C-Reativa/metabolismo , LDL-Colesterol/metabolismo , Ensaio de Imunoadsorção Enzimática , Feminino , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Glucose/análise , Hemoglobinas Glicadas/análise , Homeostase , Humanos , Inflamação , Leucócitos Mononucleares/citologia , Masculino , Pessoa de Meia-Idade , RNA Mensageiro/metabolismo , RNA Interferente Pequeno/metabolismo
10.
Acta Biochim Biophys Sin (Shanghai) ; 49(7): 567-572, 2017 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-28472221

RESUMO

Peptidylarginine deiminase 4 (PADI4), a new histone modification enzyme, which converts both arginine and monomethyl-arginine to citrulline, has gained massive attention in recent years as a potential regulator of gene transcription. Recent studies have shown that arginine residues R2, R8, R17, and R26 in the H3 tail and R3 in the H4 tail can be deiminated by PADI4. This kind of histone post-translational modification has the potential to antagonize histone methylation and coordinate with histone deacetylation to regulate gene transcription. PADI4 also deiminates non-histone proteins, such as p300, NPM1, ING4, RPS2, and DNMT3A. PADI4 has been shown to involve in cell apoptosis and differentiation. Moreover, PADI4 can interact with tumor suppressor p53 and regulate the transcriptional activity of p53. Dysregulation of PADI4 is implicated in a variety of diseases, including rheumatoid arthritis, tumor development, and multiple sclerosis. A wide variety of PADI4 inhibitors have been identified. Further understanding of PADI4 functions may lead to novel diagnostic and therapeutic approaches in these diseases. This review summarizes the recent progress in the study of the regulation mechanism of PADI4 on gene transcription and the major physiological functions of PADI4 in human diseases.


Assuntos
Citrulinação , Regulação da Expressão Gênica , Desiminases de Arginina em Proteínas/fisiologia , Acetilação , Apoptose , Artrite Reumatoide/etiologia , Biocatálise , Diferenciação Celular , Humanos , Neoplasias/etiologia , Nucleofosmina , Proteína-Arginina Desiminase do Tipo 4 , Proteína Supressora de Tumor p53/fisiologia
11.
Hum Immunol ; 78(2): 232-237, 2017 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-27816498

RESUMO

Ankylosing spondylitis (AS) is an autoimmune disease and characterized by chronic inflammatory arthritis. Tumor necrosis factor α induced protein-8 like-2 (TIPE2) is responsible for maintaining immune homeostasis by inhibiting the secretion of inflammatory cytokines in the condition of inflammation. However, whether TIPE2 participates in the development of AS remains unknown. In this study, we measured the mRNA expression of TIPE2 and TIPE1 in peripheral blood mononuclear cells (PBMCs) from 45 AS patients and 40 healthy controls by qRT-PCR. The results showed TIPE2 expression was significantly increased in AS patients compared with controls (P=0.0066), while there was no significant difference for TIPE1 between two groups (P=0.2302). Moreover, the expression of TIPE2 mRNA in AS patients were decreased after treatment with TNF-α blocker (P<0.001). In addition, we found that TNF-α or plasma from AS patients induced TIPE2 expression in THP-1 cells in vitro. More importantly, the TIPE2 mRNA expression levels were negatively correlated with TNF-α, hsCRP and bath ankylosing spondylitis disease activity index (BASDAI) (r=-0.3574, P=0.0159; r=-0.3174, P=0.0336; r=-0.6000, P<0.0001; respectively) in the AS patients. These results indicated that TIPE2 contributes to the pathogenesis of AS.


Assuntos
Peptídeos e Proteínas de Sinalização Intracelular/genética , Leucócitos Mononucleares/imunologia , Espondilite Anquilosante/genética , Adulto , Linhagem Celular , Progressão da Doença , Feminino , Homeostase , Humanos , Masculino , RNA Mensageiro/análise , Fator de Necrose Tumoral alfa/metabolismo , Regulação para Cima , Adulto Jovem
12.
Biomed Pharmacother ; 83: 936-941, 2016 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-27522256

RESUMO

Long non-coding RNAs (lncRNA) have been reported as key regulators in the progression and metastasis of prostate cancer (PCa). In this study, we found that the expression levels of HCG11 in PCa tissues were significantly lower than those in non-tumor tissues in publically available databases and in human PCa samples. Our results showed the expression levels of HCG11 in patients with PCa were associated with the age, Lymph Node Status (LN status), preoperative PSA level, Gleason score, and biochemical recurrence (BCR). Kaplan-Meier analysis indicated that downregulation of HCG11 expression in tissues was associated with poor survival of PCa patients. GO and KEGG pathway analysis were applied to explore the potential roles of HCG11. Moreover, a HCG11 mediated ceRNA network was built using co-expression relationships of the differentially expressed mRNAs and miRNAs. We believed that this study will provide a potential new therapeutic and prognostic target for prostate cancer.


Assuntos
Regulação para Baixo/genética , Regulação Neoplásica da Expressão Gênica , Neoplasias da Próstata/genética , RNA Longo não Codificante/genética , Ontologia Genética , Redes Reguladoras de Genes , Humanos , Masculino , Pessoa de Meia-Idade , Análise Multivariada , Recidiva Local de Neoplasia/patologia , Prognóstico , Neoplasias da Próstata/patologia , RNA Longo não Codificante/metabolismo , Transdução de Sinais/genética
13.
Biochimie ; 99: 215-8, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24384472

RESUMO

p53 is a transcriptional regulator in the nucleus that functions as a tumor suppressor and its mutations are frequently found in human tumors. It has been reported that p53 with R213Q mutation is exist in certain tumor cell lines and its methylation on R213 as well. However, the mechanisms and consequences of these modifications on p53 function are not fully understood. Mutations of p53 at R213Q (R/Q) and R213K (R/K) were respectively constructed and transfected into the p53 null H1299 cells. As shown in luciferase reporter assays, either R/Q or R/K disrupted the efficiency of p53 transactivation. EMSA and ChIP assays revealed that these mutants were less efficient in targeting the consensus binding sequences of p53 in the regulatory region of p21 gene. In addition, R/Q and R/K mutants attenuated the expression of p21 gene and counteracted the p53 mediated G1/S arrest to deliver a normal cell cycle progression as in the mock H1299 cells. Through this study, we have provided the first evidence on the pivotal role of arginine 213 that determines the p53 mediated functions of p21 in human cancer cells.


Assuntos
Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Mutação de Sentido Incorreto , Proteína Supressora de Tumor p53/genética , Sequência de Bases , Sítios de Ligação , Linhagem Celular Tumoral , Inibidor de Quinase Dependente de Ciclina p21/genética , Expressão Gênica , Humanos , Ligação Proteica , Ativação Transcricional , Proteína Supressora de Tumor p53/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA