Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Cancer Lett ; 511: 68-76, 2021 07 28.
Artigo em Inglês | MEDLINE | ID: mdl-33957184

RESUMO

Immune checkpoints within the tumor microenvironment (TME) play important roles in modulating host antitumor immunity. Checkpoint-based immunotherapies (e.g. immune checkpoint inhibitors) have revolutionized cancer therapy. However, there are still many drawbacks with current checkpoint immunotherapies in clinical practice, such as unresponsiveness, resistance, tumor hyperprogression, autoimmune-related adverse events, and limited efficacy with some solid malignances. These drawbacks highlight the need to further investigate the mechanisms underlying the therapeutic effects, as well as the need to identify new targets for cancer immunotherapy. With the discovery of emerging immune checkpoints in the TME, the development of strategies targeting the pivotal immunomodulators for cancer treatment has been significantly advanced in the past decade. In this review, we summarize and classify the novel emerging immune checkpoints beyond the extensively studied ones (e.g. PD-1, PD-L1, CTLA-4, LAG-3 and TIM-3) in the TME, and provide an update on the clinical trials targeting these key immune molecules.


Assuntos
Imunoterapia/métodos , Neoplasias/tratamento farmacológico , Microambiente Tumoral/imunologia , Humanos , Neoplasias/imunologia
2.
Development ; 147(17)2020 09 02.
Artigo em Inglês | MEDLINE | ID: mdl-32747435

RESUMO

Homeostasis of intestinal stem cells (ISCs) is maintained by the orchestration of niche factors and intrinsic signaling networks. Here, we have found that deletion of Erk1 and Erk2 (Erk1/2) in intestinal epithelial cells at embryonic stages resulted in an unexpected increase in cell proliferation and migration, expansion of ISCs, and formation of polyp-like structures, leading to postnatal death. Deficiency of epithelial Erk1/2 results in defects in secretory cell differentiation as well as impaired mesenchymal cell proliferation and maturation. Deletion of Erk1/2 strongly activated Wnt signaling through both cell-autonomous and non-autonomous mechanisms. In epithelial cells, Erk1/2 depletion resulted in loss of feedback regulation, leading to Ras/Raf cascade activation that transactivated Akt activity to stimulate the mTor and Wnt/ß-catenin pathways. Moreover, Erk1/2 deficiency reduced the levels of Indian hedgehog and the expression of downstream pathway components, including mesenchymal Bmp4 - a Wnt suppressor in intestines. Inhibition of mTor signaling by rapamycin partially rescued Erk1/2 depletion-induced intestinal defects and significantly prolonged the lifespan of mutant mice. These data demonstrate that Erk/Mapk signaling functions as a key modulator of Wnt signaling through coordination of epithelial-mesenchymal interactions during intestinal development.


Assuntos
Intestinos/embriologia , Sistema de Sinalização das MAP Quinases , Via de Sinalização Wnt , Animais , Proteína Morfogenética Óssea 4/genética , Proteína Morfogenética Óssea 4/metabolismo , Camundongos , Camundongos Knockout , Proteína Quinase 1 Ativada por Mitógeno/genética , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/genética , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Proteínas Proto-Oncogênicas c-akt/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , Serina-Treonina Quinases TOR/genética , Serina-Treonina Quinases TOR/metabolismo , Quinases raf/genética , Quinases raf/metabolismo
3.
Biochem Biophys Res Commun ; 519(4): 754-760, 2019 11 19.
Artigo em Inglês | MEDLINE | ID: mdl-31547988

RESUMO

Wnt signaling is essential for the maintenance of adult stem cells and its aberrant activation is a stimulator of carcinogenesis. The transmembrane protein, Wntless, is an essential Wnt signaling component through regulating the secretion of Wnt ligands. Here, we generated a mouse model with specific Wntless knockout in intestinal epithelium to study its function in the intestinal epithelium. Wntless knockout exhibits no obvious defects in mice but significantly disrupted proliferation and differentiation of small intestinal organoids. We also discovered that these deficiencies could be partially rescued by Wnt3a supplement but not Wnt9b. To further investigate the role of Wntless in tumorigenesis, APC-deficient spontaneous intestinal tumors and chemical induced colorectal cancer mouse models were employed. To our surprise, intestinal epithelium-specific knockout of Wntless did not cause significant differences in tumor number and size. In summary, our data demonstrated that epithelial Wntless was required for the growth and differentiation of small intestinal organoids but not in live animals, suggesting the other tissues, such as mesenchymal tissue, play critical role for Wnt secretion in both intestinal homeostasis as well as tumorigenesis.


Assuntos
Transformação Celular Neoplásica/genética , Modelos Animais de Doenças , Mucosa Intestinal/metabolismo , Neoplasias Intestinais/genética , Receptores Acoplados a Proteínas G/genética , Animais , Diferenciação Celular/genética , Proliferação de Células/genética , Transformação Celular Neoplásica/metabolismo , Neoplasias Intestinais/metabolismo , Camundongos Knockout , Camundongos Transgênicos , Organoides/metabolismo , Organoides/patologia , Receptores Acoplados a Proteínas G/metabolismo
4.
Int J Cancer ; 140(12): 2734-2747, 2017 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-28316092

RESUMO

The clearance of oxidative stress compounds is critical for the protection of the organism from malignancy, but how this key physiological process is regulated is not fully understood. Here, we found that the expression of GPRC5A, a well-characterized tumor suppressor in lung cancer, was elevated in colorectal cancer tissues in patients. In both cancer cell lines and a colitis-associated cancer model in mice, we found that GPRC5A deficiency reduced cell proliferation and increased cell apoptosis as well as inhibited tumorigenesis in vivo. Through RNA-Seq transcriptome analysis, we identified oxidative stress associated pathways were dysregulated. Moreover, in GPRC5A deficient cells and mouse tissues, the oxidative agents were reduced partially due to increased glutathione (GSH) level. Mechanistically, GPRC5A regulates NF-κB mediated Vanin-1 expression which is the predominant enzyme for cysteamine generation. Administration of cystamine (the disulfide form of cysteamine) in GPRC5A deficient cell lines inhibited γ-GCS activity, leading to reduction of GSH level and increase of cell growth. Taken together, our studies suggest that GPRC5a is a potential biomarker for colon cancer and promotes tumorigenesis through stimulation of Vanin-1 expression and oxidative stress in colitis associated cancer. This study revealed an unexpected oncogenic role of GPRC5A in colorectal cancer suggesting there are complicated functional and molecular mechanism differences of this gene in distinct tissues.


Assuntos
Amidoidrolases/genética , Neoplasias Colorretais/genética , Regulação Neoplásica da Expressão Gênica , Estresse Oxidativo , Receptores Acoplados a Proteínas G/genética , Animais , Apoptose/genética , Biomarcadores Tumorais/genética , Biomarcadores Tumorais/metabolismo , Western Blotting , Linhagem Celular Tumoral , Proliferação de Células/genética , Colite/complicações , Neoplasias Colorretais/complicações , Neoplasias Colorretais/metabolismo , Progressão da Doença , Proteínas Ligadas por GPI/genética , Perfilação da Expressão Gênica/métodos , Glutationa/metabolismo , Células HEK293 , Células HT29 , Humanos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptores Acoplados a Proteínas G/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Regulação para Cima
5.
J Immunol ; 195(1): 339-46, 2015 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-26026060

RESUMO

The mammalian target of rapamycin (mTOR) signaling pathway integrates environmental cues to regulate cell growth and survival through various mechanisms. However, how mTORC1 responds to acute inflammatory signals to regulate bowel regeneration is still obscure. In this study, we investigated the role of mTORC1 in acute inflammatory bowel disease. Inhibition of mTORC1 activity by rapamycin treatment or haploinsufficiency of Rheb through genetic modification in mice impaired intestinal cell proliferation and induced cell apoptosis, leading to high mortality in dextran sodium sulfate- and 2,4,6-trinitrobenzene sulfonic acid-induced colitis models. Through bone marrow transplantation, we found that mTORC1 in nonhematopoietic cells played a major role in protecting mice from colitis. Reactivation of mTORC1 activity by amino acids had a positive therapeutic effect in mTORC1-deficient Rheb(+/-) mice. Mechanistically, mTORC1 mediated IL-6-induced Stat3 activation in intestinal epithelial cells to stimulate the expression of downstream targets essential for cell proliferation and tissue regeneration. Therefore, mTORC1 signaling critically protects against inflammatory bowel disease through modulation of inflammation-induced Stat3 activity. As mTORC1 is an important therapeutic target for multiple diseases, our findings will have important implications for the clinical usage of mTORC1 inhibitors in patients with acute inflammatory bowel disease.


Assuntos
Colite/imunologia , Proteínas Monoméricas de Ligação ao GTP/imunologia , Complexos Multiproteicos/antagonistas & inibidores , Neuropeptídeos/imunologia , Fator de Transcrição STAT3/imunologia , Sirolimo/farmacologia , Serina-Treonina Quinases TOR/antagonistas & inibidores , Animais , Transplante de Medula Óssea , Células CACO-2 , Proliferação de Células/efeitos dos fármacos , Colite/induzido quimicamente , Colite/genética , Colite/mortalidade , Regulação da Expressão Gênica , Haploinsuficiência , Humanos , Interleucina-6/genética , Interleucina-6/imunologia , Alvo Mecanístico do Complexo 1 de Rapamicina , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Proteínas Monoméricas de Ligação ao GTP/deficiência , Proteínas Monoméricas de Ligação ao GTP/genética , Complexos Multiproteicos/genética , Complexos Multiproteicos/imunologia , Neuropeptídeos/deficiência , Neuropeptídeos/genética , Proteína Enriquecida em Homólogo de Ras do Encéfalo , Fator de Transcrição STAT3/genética , Transdução de Sinais , Dodecilsulfato de Sódio , Análise de Sobrevida , Serina-Treonina Quinases TOR/genética , Serina-Treonina Quinases TOR/imunologia , Ácido Trinitrobenzenossulfônico
6.
Nucleic Acids Res ; 41(11): e120, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23630316

RESUMO

Transcription activator-like effector nucleases (TALENs) are a powerful new approach for targeted gene disruption in various animal models, but little is known about their activities in Mus musculus, the widely used mammalian model organism. Here, we report that direct injection of in vitro transcribed messenger RNA of TALEN pairs into mouse zygotes induced somatic mutations, which were stably passed to the next generation through germ-line transmission. With one TALEN pair constructed for each of 10 target genes, mutant F0 mice for each gene were obtained with the mutation rate ranged from 13 to 67% and an average of ∼40% of total healthy newborns with no significant differences between C57BL/6 and FVB/N genetic background. One TALEN pair with single mismatch to their intended target sequence in each side failed to yield any mutation. Furthermore, highly efficient germ-line transmission was obtained, as all the F0 founders tested transmitted the mutations to F1 mice. In addition, we also observed that one bi-allele mutant founder of Lepr gene, encoding Leptin receptor, had similar diabetic phenotype as db/db mouse. Together, our results suggest that TALENs are an effective genetic tool for rapid gene disruption with high efficiency and heritability in mouse with distinct genetic background.


Assuntos
Endodesoxirribonucleases/metabolismo , Marcação de Genes/métodos , Animais , Proteínas de Ligação a DNA/genética , Endodesoxirribonucleases/genética , Mutação INDEL , Camundongos , Taxa de Mutação , Fenótipo
7.
J Biol Chem ; 288(13): 8794-803; discussion 8804, 2013 Mar 29.
Artigo em Inglês | MEDLINE | ID: mdl-23393138

RESUMO

Lgr4/Gpr48 is one of the newly identified R-spondins receptors and potentiates Wnt signaling, which regulates intestinal homeostasis. We used a hypomorphic mouse strain to determine the role of Lgr4 in intestinal inflammation and recovery. Intestinal inflammation was induced with dextran sulfate sodium (DSS) followed by a recovery period. Intestinal inflammation symptoms and molecular mechanisms were examined. We found that Lgr4(-/-) mice exhibited dramatically higher susceptibility to and mortality from DSS-induced inflammatory bowel disease than WT mice. Lgr4 deficiency resulted in greatly reduced numbers of either Paneth cells or stem cells in the intestine. During the intestinal regeneration process, cell proliferation but not apoptosis of intestinal epithelial cells was significantly impaired in Lgr4(-/-) mice. When Wnt/ß-catenin signaling was reactivated by crossing with APC(min)(/+) mice or by treating with a GSK-3ß inhibitor, the number of Paneth cells was partially restored and the mortality caused by DSS-induced inflammatory bowel disease was strikingly reduced in Lgr4-deficient animals. Thus, Lgr4 is critically involved in the maintenance of intestinal homeostasis and protection against inflammatory bowel disease through modulation of the Wnt/ß-catenin signaling pathway.


Assuntos
Colite/metabolismo , Dextranos/farmacologia , Regulação da Expressão Gênica , Predisposição Genética para Doença , Doenças Inflamatórias Intestinais/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Sulfatos/farmacologia , Animais , Transplante de Medula Óssea , Colite/genética , Descoberta de Drogas , Deleção de Genes , Homeostase , Doenças Inflamatórias Intestinais/genética , Camundongos , Camundongos Transgênicos , Receptores Acoplados a Proteínas G/genética , Regeneração , Transdução de Sinais , Células-Tronco/citologia , Trombospondinas/metabolismo , Proteínas Wnt/metabolismo , beta Catenina/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...