Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
1.
Genes Dis ; 10(4): 1429-1444, 2023 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-37397521

RESUMO

DNA is highly vulnerable to spontaneous and environmental timely damage in living cells. DNA damage may cause genetic instability and increase cancer risk if the damages are not repaired timely and efficiently. Human cells possess several DNA damage response (DDR) mechanisms to protect the integrity of their genome. Clarification of the mechanisms underlying the DNA damage response following lethal damage will facilitate the identification of therapeutic targets for cancers. Histone post-translational modifications (PTMs) have been indicated to play different roles in the repair of DNA damage. In this context, histone PTMs regulate recruitment of downstream effectors, and facilitate appropriate repair response. This review outlines the current understanding of different histone PTMs in response to DNA damage repair, besides, enumerates the role of new type PTMs such as histone succinylation and crotonylation in regulating DNA damage repair processes.

2.
J Plant Physiol ; 284: 153961, 2023 May.
Artigo em Inglês | MEDLINE | ID: mdl-36933340

RESUMO

The tubers of the widely distributed Cyperus esculentus are rich in oil, and therefore, the plant is considered to have a high utilization value in the vegetable oil industry. Oleosins and caleosins are lipid-associated proteins found in oil bodies of seeds; however oleosins and caleosins genes have not been identified in C. esculentus. In this study, we performed transcriptome sequencing and lipid metabolome analysis of C. esculentus tubers at four developmental stages to obtain the information on their genetic profile, expression trends, and metabolites in oil accumulation pathways. Overall, 120,881 non-redundant unigenes and 255 lipids were detected; 18 genes belonged to the acetyl-CoA carboxylase (ACC), malonyl-CoA:ACP transacylase (MCAT), ß-ketoacyl-ACP synthase (KAS), and fatty acyl-ACP thioesterase (FAT) gene families involved in fatty acid biosynthesis, and 16 genes belonged to the glycerol-3-phosphate acyltransferase (GPAT), diacylglycerol acyltransferase 3 (DGAT3), phospholipid:diacylglycerol acyltransferase (PDAT), FAD2, and lysophosphatidic acid acyltransferase (LPAAT) gene families playing important roles in triacylglycerol synthesis. We also identified 9 oleosin- and 21 caleosin-encoding genes in C. esculentus tubers. These results provide detailed information on the C. esculentus transcriptional and metabolic profiles, which can be used as reference for the development of strategies to increase oil content in C. esculentus tubers.


Assuntos
Cyperus , Diacilglicerol O-Aciltransferase , Diacilglicerol O-Aciltransferase/genética , Cyperus/genética , Cyperus/metabolismo , Proteínas de Plantas/metabolismo , Sementes/metabolismo , Óleos de Plantas/metabolismo
3.
J Oncol ; 2022: 7319641, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35847360

RESUMO

Aim: To explore the role of Rac1 on sorafenib resistance in hepatocellular carcinoma. Methods: CCK-8, wound healing assay, Transwell, and cell cycle assay were used to detect the tumor cells development. Cell viability was assessed by MTT. The glycolytic pathway was revealed by cellular metabolism assays. Result: We recovered that Rac1 upregulation was related to HCC patients' poorer prognosis. Forced expression of Rac1 promoted cell development and sorafenib chemoresistance in HCC cells. Rac1 inhibitor EHop-016 and sorafenib combination markedly prevented cell viability, G2/M phase cycle arrest, and apoptosis than single therapy. Furthermore, combination therapy decreased glycolysis in HCC cells. In vivo, the tumor growth was significantly prevented by combination therapy single therapy. Conclusion: Our research declares that Rac1 inhibition could block sorafenib resistance in HCC by decreasing glycolysis, which would provide an underlying target for HCC therapy.

4.
Proc Natl Acad Sci U S A ; 119(29): e2205574119, 2022 07 19.
Artigo em Inglês | MEDLINE | ID: mdl-35858331

RESUMO

Intestinal barrier immunity is essential for controlling gut microbiota without eliciting harmful immune responses, while its defect contributes to the breakdown of intestinal homeostasis and colitis development. Chemerin, which is abundantly expressed in barrier tissues, has been demonstrated to regulate tissue inflammation via CMKLR1, its functional receptor. Several studies have reported the association between increased expression of chemerin-CMKLR1 and disease severity and immunotherapy resistance in inflammatory bowel disease (IBD) patients. However, the pathophysiological role of endogenous chemerin-CMKLR1 signaling in intestinal homeostasis remains elusive. We herein demonstrated that deficiency of chemerin or intestinal epithelial cell (IEC)-specific CMKLR1 conferred high susceptibility to microbiota-driven neutrophilic colon inflammation and subsequent tumorigenesis in mice following epithelial injury. Unexpectedly, we found that lack of chemerin-CMKLR1 signaling specifically reduced expression of lactoperoxidase (LPO), a peroxidase that is predominantly expressed in colonic ECs and utilizes H2O2 to oxidize thiocyanates to the antibiotic compound, thereby leading to the outgrowth and mucosal invasion of gram-negative bacteria and dysregulated CXCL1/2-mediated neutrophilia. Importantly, decreased LPO expression was causally linked to aggravated microbiota-driven colitis and associated tumorigenesis, as LPO supplementation could completely rescue such phenotypes in mice deficient in epithelial chemerin-CMKLR1 signaling. Moreover, epithelial chemerin-CMKLR1 signaling is necessary for early host defense against bacterial infection in an LPO-dependent manner. Collectively, our study reveals that the chemerin-CMKLR1/LPO axis represents an unrecognized immune mechanism that potentiates epithelial antimicrobial defense and restricts harmful colonic neutrophilia and suggests that LPO supplementation may be beneficial for microbiota dysbiosis in IBD patients with a defective innate antimicrobial mechanism.


Assuntos
Carcinogênese , Quimiocinas , Colite , Colo , Microbioma Gastrointestinal , Peptídeos e Proteínas de Sinalização Intercelular , Lactoperoxidase , Receptores de Quimiocinas , Animais , Carcinogênese/imunologia , Transformação Celular Neoplásica , Quimiocinas/genética , Quimiocinas/metabolismo , Colite/imunologia , Colite/microbiologia , Colo/imunologia , Colo/microbiologia , Peróxido de Hidrogênio/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/genética , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiologia , Lactoperoxidase/metabolismo , Camundongos , Neutrófilos/imunologia , Receptores de Quimiocinas/genética , Receptores de Quimiocinas/metabolismo
5.
Hepatology ; 75(1): 28-42, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34387870

RESUMO

BACKGROUND AND AIMS: We previously demonstrated that cancer-associated fibroblasts (CAFs) promote tumor growth through recruitment of myeloid-derived suppressor cells (MDSCs). 5-lipoxygenase (5-LO) is highly expressed in myeloid cells and is critical for synthesizing leukotriene B4 (LTB4), which is involved in tumor progression by activating its receptor leukotriene B4 receptor type 2 (BLT2). In this study, we investigated whether and how CAFs regulate MDSC function to enhance cancer stemness, the driving force of the cancer aggressiveness and chemotherapy refractoriness, in highly desmoplastic intrahepatic cholangiocarcinoma (ICC). APPROACH AND RESULTS: RNA-sequencing analysis revealed enriched metabolic pathways but decreased inflammatory pathways in cancer MDSCs compared with blood MDSCs from patients with ICC. Co-injection of ICC patient-derived CAFs promoted cancer stemness in an orthotopic ICC model, which was blunted by MDSC depletion. Conditioned media (CM) from CAF-educated MDSCs drastically promoted tumorsphere formation efficiency and stemness marker gene expression in ICC cells. CAF-CM stimulation increased expression and activity of 5-LO in MDSCs, while 5-LO inhibitor impaired the stemness-enhancing capacity of MDSCs in vitro and in vivo. Furthermore, IL-6 and IL-33 primarily expressed by CAFs mediated hyperactivated 5-LO metabolism in MDSCs. We identified the LTB4-BLT2 axis as the critical downstream metabolite signaling of 5-LO in promoting cancer stemness, as treatment with LTB4 was elevated in CAF-educated MDSCs, or blockade of BLT2 (which was preferentially expressed in stem-like ICC cells) significantly reduced stemness-enhancing effects of CAF-educated MDSCs. Finally, BLT2 blockade augmented chemotherapeutic efficacy in ICC patient-derived xenograft models. CONCLUSIONS: Our study reveals a role for CAFs in orchestrating the optimal cancer stemness-enhancing microenvironment by educating MDSCs, and suggests the 5-LO/LTB4-BLT2 axis as promising therapeutic targets for ICC chemoresistance by targeting cancer stemness.


Assuntos
Araquidonato 5-Lipoxigenase/metabolismo , Neoplasias dos Ductos Biliares/patologia , Fibroblastos Associados a Câncer/metabolismo , Colangiocarcinoma/patologia , Células-Tronco Neoplásicas/patologia , Animais , Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Neoplasias dos Ductos Biliares/tratamento farmacológico , Ductos Biliares Intra-Hepáticos/patologia , Comunicação Celular , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Colangiocarcinoma/tratamento farmacológico , Meios de Cultivo Condicionados/metabolismo , Resistencia a Medicamentos Antineoplásicos , Humanos , Inibidores de Lipoxigenase/farmacologia , Masculino , Camundongos , Células Supressoras Mieloides/metabolismo , Células-Tronco Neoplásicas/efeitos dos fármacos , Receptores do Leucotrieno B4/antagonistas & inibidores , Receptores do Leucotrieno B4/metabolismo , Microambiente Tumoral/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto
6.
J Leukoc Biol ; 110(2): 293-300, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-34184320

RESUMO

Chronic pancreatitis (CP) is a continuing or relapsing inflammatory disease of the pancreas, characterized by fibrosis of the whole tissue. The regulatory mechanisms of the immune microenvironment in the pathogenesis of CP are still not clear. Immune cells, especially myeloid cells, play an important role in the pathogenesis of pancreatitis. Understanding the regulatory mechanisms of immune infiltration has a significant impact on CP intervention. Here, we demonstrated that transcription factor STAT5 was involved in and critical for the progression of CP. Inflammatory stress could significantly increase the expression and activation of STAT5 during CP. STAT5 deficiency or inhibition contributed to alleviating pancreatic inflammation and fibrosis in CP mice. The increased neutrophil infiltration, mediated by up-regulated GM-CSF, was responsible for the pancreatitis-promoting activity of STAT5. Our investigation highlighted the importance of STAT5 in regulating the immune microenvironment of CP. Targeting STAT5 may hold distinct promise for clinical treatment to alleviate CP.


Assuntos
Fator Estimulador de Colônias de Granulócitos e Macrófagos/metabolismo , Neutrófilos/imunologia , Neutrófilos/metabolismo , Pancreatite Crônica/etiologia , Pancreatite Crônica/metabolismo , Fator de Transcrição STAT5/metabolismo , Animais , Biomarcadores , Suscetibilidade a Doenças , Fibrose , Humanos , Pancreatite Crônica/patologia , Estresse Fisiológico
7.
Cell Death Dis ; 12(3): 273, 2021 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-33723230

RESUMO

Chronic pancreatitis (CP) is characterized by a wide range of irreversible fibro-inflammatory diseases with largely ambiguous pathogenesis. Although neddylation pathway has been implicated in regulating immune responses, whether the dysregulation of neddylation is involved in the progression of CP and how neddylation regulates the inflammatory microenvironment of CP have not yet been reported. Here, we demonstrate that global inactivation of neddylation pathway by MLN4924 significantly exacerbates chronic pancreatitis. The increased M2 macrophage infiltration, mediated by the upregulated chemokine (C-C motif) ligand 5 (CCL5), is responsible for the enhanced pancreatitis-promoting activity of MLN4924. Both CCL5 blockade and macrophage depletion contribute to alleviating pancreatic fibrosis and inflammation in MLN4924-treated CP mice. Mechanistic investigation identifies that inactivation of Cullin-RING ligases (CRLs) stabilizes cellular levels of hypoxia-inducible factor 1α (HIF-1α), which increases CCL5 expression by promoting CCL5 transactivation. Clinically, UBE2M expression remarkably decreases in human CP tissues compared with normal specimens and the levels of CCL5 and M2 marker CD163 are negatively correlated with UBE2M intensity, suggesting that neddylation is involved in the pathogenesis of pancreatitis. Hence, our studies reveal a neddylation-associated immunopathogenesis of chronic pancreatitis and provide new ideas for the disease treatment.


Assuntos
Quimiocina CCL5/metabolismo , Quimiotaxia , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Macrófagos/metabolismo , Pâncreas/metabolismo , Pancreatite Crônica/metabolismo , Enzimas Ativadoras de Ubiquitina/metabolismo , Animais , Linhagem Celular , Quimiocina CCL5/genética , Quimiotaxia/efeitos dos fármacos , Ciclopentanos/toxicidade , Modelos Animais de Doenças , Inibidores Enzimáticos/toxicidade , Feminino , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Macrófagos/efeitos dos fármacos , Macrófagos/patologia , Camundongos Endogâmicos C57BL , Pâncreas/efeitos dos fármacos , Pâncreas/patologia , Pancreatite Crônica/induzido quimicamente , Pancreatite Crônica/genética , Pancreatite Crônica/patologia , Fenótipo , Pirimidinas/toxicidade , Transdução de Sinais , Enzimas Ativadoras de Ubiquitina/antagonistas & inibidores , Ubiquitinação
8.
Cancer Immunol Res ; 8(11): 1440-1451, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32917658

RESUMO

The mechanisms responsible for radioresistance in pancreatic cancer have yet to be elucidated, and the suppressive tumor immune microenvironment must be considered. We investigated whether the radiotherapy-augmented Warburg effect helped myeloid cells acquire an immunosuppressive phenotype, resulting in limited treatment efficacy of pancreatic ductal adenocarcinoma (PDAC). Radiotherapy enhanced the tumor-promoting activity of myeloid-derived suppressor cells (MDSC) in pancreatic cancer. Sustained increase in lactate secretion, resulting from the radiation-augmented Warburg effect, was responsible for the enhanced immunosuppressive phenotype of MDSCs after radiotherapy. Hypoxia-inducible factor-1α (HIF-1α) was essential for tumor cell metabolism and lactate-regulated activation of MDSCs via the G protein-coupled receptor 81 (GPR81)/mTOR/HIF-1α/STAT3 pathway. Blocking lactate production in tumor cells or deleting Hif-1α in MDSCs reverted antitumor T-cell responses and effectively inhibited tumor progression after radiotherapy in pancreatic cancer. Our investigation highlighted the importance of radiation-induced lactate in regulating the inhibitory immune microenvironment of PDAC. Targeting lactate derived from tumor cells and the HIF-1α signaling in MDSCs may hold distinct promise for clinical therapies to alleviate radioresistance in PDAC.


Assuntos
Terapia de Imunossupressão/métodos , Ácido Láctico/metabolismo , Células Supressoras Mieloides/imunologia , Neoplasias Pancreáticas/imunologia , Animais , Modelos Animais de Doenças , Humanos , Camundongos
9.
Immun Inflamm Dis ; 8(4): 782-792, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-32749072

RESUMO

INTRODUCTION: Protein neddylation, one of the most important posttranslational modifications that tagging neuronal precursor cell-expressed developmentally downregulated protein 8 onto substrate proteins, plays fundamental roles in the process of many cellular functions. A number of studies have demonstrated the critical roles of neddylation modification in multiple pathophysiological processes, but its regulatory role in the immune system has only been finitely unveiled. METHODS: In this review, the latest advances in the field of neddylation modification in regulating the immune responses are succinctly discussed. RESULTS: Neddylation modification acts as a crucial modulator of innate immune cells (neutrophils, macrophages, and dendritic cells) and lymphocytes. Dysregulation of neddylation alters characteristics and functions of those cells due to abnormal degradation of key signaling molecules involved in immunoregulation. Furthermore, the ectopic immune responses caused by the abnormal neddylation play pivotal roles in a variety of immune-related diseases, such as infection, inflammation, and cancer. CONCLUSIONS: The pivotal roles of neddylation pathway in immunoregulation are attracted more and more attention, which may provide new insights into the pathogenesis of a variety of immune-related diseases and help to indicate new therapeutic targets and potential treatment strategies.


Assuntos
Imunidade , Humanos , Proteína NEDD8 , Neoplasias , Transdução de Sinais
10.
J Cancer ; 10(26): 6673-6680, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31777596

RESUMO

Emerging evidence revealed the critical role of systematic inflammation in pancreatic ductal adenocarcinoma (PDAC). In the present study, we reviewed the records of 279 patients with advanced PDAC. Among them, 147 cases were used as the training cohort and another 132 as the validation cohort. In the training cohort, distant metastasis, carbohydrate antigen 19-9 (CA19-9), Glasgow prognostic score (GPS), neutrophil-to-lymphocyte ratio (NLR), and lymphocyte-to-monocyte ratio (LMR) were independent prognostic factors in Cox regression. A nomogram based on these factors was generated to predict median survival time and survival probabilities at 6, 12, and 18 months. The nomogram showed a better discriminatory ability than the American Joint Committee on Cancer (AJCC) TNM staging (C-index: 0.727 vs. 0.610). In the validation cohort, a nomogram composed of the same variables also showed a high discriminatory ability (C-index: 0.784). In the low-risk group with a nomogram total point (NTP) value of more than 175, patients receiving combination therapy showed better prognosis than those receiving monotherapy (P=0.015). In conclusion, the nomogram based on inflammatory biomarkers can serve as useful prognostic tool for advanced PDAC. In addition, patients with high NTP can greater benefit from combination chemotherapy than monotherapy.

11.
Neoplasia ; 21(12): 1133-1142, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31759251

RESUMO

Desmoplasia is a hallmark of intrahepatic cholangiocarcinoma (ICC), which constitutes a barrier to infiltration of lymphocyte, but not myeloid cells. Given that dense desmoplastic stroma has been reported to be a barrier to infiltration of lymphocyte, but not myeloid cells. We here investigated whether fibroblastic FAP influenced ICC progression via non-T cell-related immune mechanisms. We demonstrated fibroblastic FAP expression was critical for STAT3 activation and CCL2 production, and ICC-CAFs were the primary source of CCL2 in human ICC microenvironment by using ICC-Fbs from six ICC patients. Fibroblastic knockdown of FAP significantly impaired the ability of ICC-CAFs to promote ICC growth, MDSCs infiltration and angiogenesis, which was restored by adding exogenous CCL2. Furthermore, interestingly, the tumor-promoting effect of fibroblastic FAP is dependent on MDSCs via secretion of CCL2, as depletion of Gr-1+ cells reversed the restoring effects of exogenous CCL2 on tumor growth and angiogenesis. In vitro migration assay confirmed that exogenous CCL2 could rescue the impaired ability of ICC-Fbs to attract Gr-1+ cells caused by fibroblastic FAP knockdown. In contrast, fibroblastic FAP knockdown had no effect on ICC cell proliferation and apoptotic resistance. Depletion MDSCs by anti-Gr-1 monoclonal antibody in subcutaneous transplanted tumor model abrogated tumor promotion by ICC-CAFs suggested that the pro-tumor function of Fibroblastic FAP relied on MDSCs. Mechanical, flow cytometry and chamber migration assay were conducted to find Fibroblastic FAP was required by the ability of ICC-CAFs to promote MDSC migration directly. Moreover, fibroblastic FAP knockdown had no effect on cell proliferation and apoptotic resistance. Here, we revealed the T-cell independent mechanisms underlying the ICC-promoting effect of fibroblastic FAP by attracting MDSCs via CCL2, which was mainly attributed to the ability of FAP to attract MDSCs and suggests that specific targeting fibroblastic FAP may represent a promising therapeutic strategy against ICC.


Assuntos
Neoplasias dos Ductos Biliares/etiologia , Neoplasias dos Ductos Biliares/metabolismo , Colangiocarcinoma/etiologia , Colangiocarcinoma/metabolismo , Gelatinases/genética , Proteínas de Membrana/genética , Células Supressoras Mieloides/metabolismo , Serina Endopeptidases/genética , Animais , Antígenos Ly/metabolismo , Apoptose/genética , Neoplasias dos Ductos Biliares/patologia , Linhagem Celular Tumoral , Movimento Celular/genética , Proliferação de Células , Colangiocarcinoma/patologia , Modelos Animais de Doenças , Endopeptidases , Fibroblastos/metabolismo , Gelatinases/metabolismo , Técnicas de Silenciamento de Genes , Xenoenxertos , Humanos , Masculino , Proteínas de Membrana/metabolismo , Camundongos , Células Supressoras Mieloides/imunologia , Fator de Transcrição STAT3/metabolismo , Serina Endopeptidases/metabolismo , Microambiente Tumoral/genética
12.
Mucosal Immunol ; 12(6): 1304-1315, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31534167

RESUMO

Enhanced gut permeability due to dysregulated epithelial tight junction is often associated with inflammatory bowel diseases (IBD), which have a greater risk for developing colorectal cancer. STAT6 activation was detected in inflamed colonic epithelium of active IBD patients, suggesting a role of epithelial STAT6 in colitis development. Here, we demonstrated that non-hematopoietic STAT6, but not hematopoietic STAT6, triggered DSS-induced colitis and subsequent tumorigenesis. This could be due to the enhancing-effect of STAT6 on gut permeability and microbiota translocation via interruption of epithelial tight junction integrity. Mechanistically, long-myosin light-chain kinase (MLCK1) was identified as a target of STAT6, leading to epithelial tight junction dysfunction and microbiota-driven colitis. Furthermore, neutralization of IL-13, which was primarily derived from type 2 innate lymphoid cells (ILC2) in a microbiota-dependent way, inhibited epithelial STAT6 activation and improved gut permeability and DSS-induced colitis. Importantly, pharmacological inhibition of STAT6 reduces murine intestinal tumor formation, and tumoral p-STAT6 levels positively correlated to the clinical stage and poor prognosis of human colorectal cancer. Thus, our study reveals a direct role of STAT6 in the disruption of epithelial tight junction integrity and colitis development, and suggests STAT6 as a potential therapeutic and prophylactic target for IBD and colitis-associated cancer.


Assuntos
Colite/metabolismo , Colo/metabolismo , Neoplasias do Colo/metabolismo , Mucosa Intestinal/metabolismo , Fator de Transcrição STAT6/metabolismo , Junções Íntimas/metabolismo , Animais , Translocação Bacteriana , Células CACO-2 , Colite/genética , Colite/microbiologia , Colite/patologia , Colo/efeitos dos fármacos , Colo/microbiologia , Colo/patologia , Neoplasias do Colo/microbiologia , Neoplasias do Colo/patologia , Neoplasias do Colo/prevenção & controle , Modelos Animais de Doenças , Impedância Elétrica , Microbioma Gastrointestinal , Genes APC , Humanos , Interleucina-13/metabolismo , Mucosa Intestinal/efeitos dos fármacos , Mucosa Intestinal/microbiologia , Mucosa Intestinal/patologia , Linfócitos/metabolismo , Linfócitos/patologia , Camundongos da Linhagem 129 , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Quinase de Cadeia Leve de Miosina/genética , Quinase de Cadeia Leve de Miosina/metabolismo , Permeabilidade , Fosforilação , Pirimidinas/farmacologia , Fator de Transcrição STAT6/antagonistas & inibidores , Fator de Transcrição STAT6/deficiência , Fator de Transcrição STAT6/genética , Transdução de Sinais , Junções Íntimas/efeitos dos fármacos , Junções Íntimas/microbiologia , Junções Íntimas/patologia , Técnicas de Cultura de Tecidos
13.
Oncogene ; 38(29): 5792-5804, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-31243299

RESUMO

Tumor-associated macrophages (TAMs) are the most abundant cancer stromal cells and play an essential role in tumor immunosuppression, providing a suitable microenvironment for cancer development and progression. However, mechanisms of regulating TAMs infiltration in tumor sites are not fully understood. Here, we show that inactivation of neddylation pathway significantly inhibits infiltration of TAMs, leading to the suppression of lung cancer metastasis. RNA-sequencing analysis revealed that neddylation inactivation suppresses the transactivation of chemotactic cytokine ligand 2 (CCL2). Mechanistically, neddylation inactivation inhibits the activity of Cullin-RING ligases (CRLs) and induces the accumulation of its substrate IκBα to block NF-κB transcriptional activity and CCL2 transactivation. As a result, neddylation inactivation exhibits lower chemotaxis of monocytes, thereby decreasing TAMs infiltration, which can be alleviated by CCL2 addition. Moreover, the expression level of NEDD8 is positively correlated with high CCL2 expression in lung adenocarcinoma, conferring a worse overall patient survival. Together, neddylation pathway promotes CCL2 transactivation and TAMs infiltration in lung cancer to provide a tumor-promoting microenvironment, which validates neddylation pathway as a promising target for anti-TAMs therapeutic strategies.


Assuntos
Adenocarcinoma/patologia , Quimiocina CCL2/metabolismo , Neoplasias Pulmonares/patologia , Macrófagos/patologia , Proteína NEDD8/metabolismo , NF-kappa B/metabolismo , Transdução de Sinais , Adenocarcinoma/metabolismo , Animais , Linhagem Celular Tumoral , Quimiocina CCL2/genética , Humanos , Neoplasias Pulmonares/metabolismo , Camundongos , Proteína NEDD8/genética , Microambiente Tumoral
14.
Cell Biol Toxicol ; 35(3): 233-245, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-31140025

RESUMO

Metastasis is the leading cause of tumor-related death from lung cancer. However, limited success has been achieved in the treatment of lung cancer metastasis due to the lack of understanding of the mechanisms that underlie the metastatic process. In this study, Lewis lung carcinoma (LLC) cells which expressed green fluorescent protein in the nucleus and red fluorescent protein in the cytoplasm were used to record metastatic process in real-time via a whole-mouse imaging system. Using this system, we show the neddylation inhibitor MLN4924 inhibits multiple steps of the metastatic process, including intravascular survival, extravasation, and formation of metastatic colonies, thus finally suppressing tumor metastasis. Mechanistically, MLN4924 efficiently inhibits the expression of MMP2, MMP9, and vimentin and disrupts the actin cytoskeleton at an early stage to impair invasive potential and subsequently causes a DNA damage response, cell cycle arrest, and apoptosis upon long exposure to MLN4924. Furthermore, MMP2 and MMP9 are overexpressed in patient lung adenocarcinoma, which conferred a worse overall survival. Together, targeting the neddylation pathway via MLN4924 suppresses multiple steps of the metastatic process, highlighting the potential therapeutic value of MLN4924 for the treatment of metastatic lung cancer.


Assuntos
Neoplasias Pulmonares/metabolismo , Proteína NEDD8/metabolismo , Metástase Neoplásica/prevenção & controle , Animais , Apoptose/fisiologia , Pontos de Checagem do Ciclo Celular , Linhagem Celular Tumoral , Movimento Celular/fisiologia , Proliferação de Células/fisiologia , Ciclopentanos/farmacologia , Regulação Neoplásica da Expressão Gênica/genética , Humanos , Neoplasias Pulmonares/fisiopatologia , Metaloproteinase 2 da Matriz/metabolismo , Metaloproteinase 9 da Matriz/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Proteína NEDD8/fisiologia , Invasividade Neoplásica/fisiopatologia , Metástase Neoplásica/fisiopatologia , Processamento de Proteína Pós-Traducional/fisiologia , Pirimidinas/farmacologia , Transdução de Sinais , Enzimas Ativadoras de Ubiquitina/metabolismo , Vimentina/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
15.
Cancer Med ; 8(3): 1326-1335, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30767430

RESUMO

Naïve and memory T cells play a pivotal role in solid tumor pathogenesis but their role in pancreatic cancer progression remains elusive. Thus, we aimed to investigate their clinical potential in advanced pancreatic cancer (APC). Flow cytometry was performed to evaluate the level of baseline peripheral naïve and memory T cells from 137 APC patients before receiving first-line chemotherapy. Interrelationships between naïve, memory T cells and clinicopathological variables were evaluated using Pearson's correlation. The prognostic impact of naïve and memory T cells were assessed by Kaplan-Meier analysis and Cox regression. The correlation between naïve/memory T cells and tumor progression was investigated by Student's t test. CD4+ naïve/memory ratio showed close correlations with hemoglobin, red blood cell (RBC), absolute neutrophil count (ANC) and platelet while CD8+ naïve/memory ratio was correlated with hemoglobin, RBC and CEA. Higher baseline lever of CD4+ CD45RO+ /CD4+ was correlated with better overall survival (OS) (P = 0.036). Patients with CD4+ naïve/memory ratio ≥0.36 had a poorer OS than those with CD4+ naïve/memory ratio <0.36 (P = 0.021). In addition, CD4+ naïve/memory ratio showed independent prognostic impact (HR 1.427, 95% CI 1.033-1.973, P = 0.031). Furthermore, poorer clinical response was correlated with higher level of CD8+ naïve/memory ratio after the third cycle of chemotherapy (P = 0.01). Besides, patients with a lower level of CD8+ naïve/memory ratio had longer progression-free survival (PFS) (P = 0.028). We propose CD4+ naïve/memory ratio as a novel prognostic biomarker for APC. In addition, CD8+ naïve/memory ratio can be a candidate marker for predicting PFS and the change of its level may reflect the progression of APC.


Assuntos
Memória Imunológica , Antígenos Comuns de Leucócito/metabolismo , Neoplasias Pancreáticas/etiologia , Neoplasias Pancreáticas/metabolismo , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/metabolismo , Adulto , Idoso , Idoso de 80 Anos ou mais , Biomarcadores , Progressão da Doença , Feminino , Humanos , Imunofenotipagem , Estimativa de Kaplan-Meier , Contagem de Linfócitos , Masculino , Pessoa de Meia-Idade , Metástase Neoplásica , Estadiamento de Neoplasias , Neoplasias Pancreáticas/mortalidade , Neoplasias Pancreáticas/patologia , Prognóstico
16.
Kidney Int ; 92(1): 89-100, 2017 07.
Artigo em Inglês | MEDLINE | ID: mdl-28318626

RESUMO

Cisplatin is an effective chemotherapeutic agent and widely used in treatment of various solid organ malignancies, including head and neck, ovarian, and testicular cancers. However, the induction of acute kidney injury (AKI) is one of its main side effects. Leukotriene B4 receptor 1 (BLT1) mediates the majority of physiological effects of leukotriene B4 (LTB4), a potent lipid chemoattractant generated at inflammation sites, but the role of the LTB4-BLT1 axis in cisplatin-induced AKI remains unknown. Here we found upregulated LTB4 synthesis and BLT1 expression in the kidney after cisplatin administration. Cisplatin was found to directly upregulate gene expression of leukotriene A4 hydrolase and stimulate LTB4 production in renal tubular epithelial cells. Reduced kidney structural/functional damage, inflammation, and apoptosis were observed in BLT1-/- mice, as well as in wild-type mice treated with the LTA4H inhibitor SC-57461A and the BLT1 antagonist U-75302. Neutrophils were likely the target of this pathway, as BLT1 absence induced a significant decrease in infiltrating neutrophils in the kidney. Adoptive transfer of neutrophils from wild-type mice restored kidney injury in BLT1-/- mice following cisplatin challenge. Thus, the LTB4-BLT1 axis contributes to cisplatin-induced AKI by mediating kidney recruitment of neutrophils, which induce inflammation and apoptosis in the kidney. Hence, the LTB4-BLT1 axis could be a potential therapeutic target in cisplatin-induced AKI.


Assuntos
Injúria Renal Aguda/metabolismo , Quimiotaxia de Leucócito , Cisplatino , Rim/metabolismo , Leucotrieno B4/metabolismo , Infiltração de Neutrófilos , Neutrófilos/metabolismo , Receptores do Leucotrieno B4/metabolismo , Injúria Renal Aguda/induzido quimicamente , Injúria Renal Aguda/patologia , Injúria Renal Aguda/prevenção & controle , Transferência Adotiva , Animais , Apoptose , Linhagem Celular , Quimiotaxia de Leucócito/efeitos dos fármacos , Modelos Animais de Doenças , Inibidores Enzimáticos/farmacologia , Epóxido Hidrolases/antagonistas & inibidores , Epóxido Hidrolases/metabolismo , Predisposição Genética para Doença , Humanos , Rim/efeitos dos fármacos , Rim/patologia , Antagonistas de Leucotrienos/farmacologia , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Infiltração de Neutrófilos/efeitos dos fármacos , Neutrófilos/efeitos dos fármacos , Neutrófilos/patologia , Neutrófilos/transplante , Fenótipo , Receptores do Leucotrieno B4/antagonistas & inibidores , Receptores do Leucotrieno B4/deficiência , Receptores do Leucotrieno B4/genética , Transdução de Sinais , Fatores de Tempo
17.
Cancer Lett ; 385: 12-20, 2017 01 28.
Artigo em Inglês | MEDLINE | ID: mdl-27826040

RESUMO

Vascular recovery or re-angiogenesis after radiotherapy plays a significant role in tumor recurrence, whereas molecular mechanisms of this process remain elusive. In this work, we found that dying glioma cells promoted post-irradiation angiogenesis through a caspase 3 dependent mechanism. Evidence in vitro and in vivo indicated that caspase 3 inhibition undermined proangiogenic effects of dying glioma cells. Proteolytic inactivation of caspase 3 in glioma cells reduced tumorigenicity. Importantly, we identified that NF-κB/COX-2/PGE2 axis acted as downstream signaling of caspase 3, mediating proangiogenic response after irradiation. Additionally, VEGF-A, regulated by caspase 3 possibly through phosphorylated eIF4E, was recognized as another downstream factor participating in the proangiogenic response. In conclusion, these data demonstrated that caspase 3 in dying glioma cells supported the proangiogenic response after irradiation by governing NF-κB/COX-2/PGE2 axis and p-eIF4E/VEGF-A signaling. While inducing caspase 3 activation has been a generally-adopted notion in cancer therapeutics, our study counterintuitively illustrated that caspase 3 activation in dying glioma cells unfavorably supported post-irradiation angiogenesis. This double-edged role of caspase 3 suggested that taming caspase 3 from the opposite side, not always activating it, may provide novel therapeutic strategies due to restricted post-irradiation angiogenesis.


Assuntos
Neoplasias Encefálicas/radioterapia , Caspase 3/metabolismo , Glioma/radioterapia , Neovascularização Patológica , Microambiente Tumoral , Inibidores da Angiogênese/farmacologia , Animais , Neoplasias Encefálicas/irrigação sanguínea , Neoplasias Encefálicas/enzimologia , Neoplasias Encefálicas/patologia , Caspase 3/genética , Inibidores de Caspase/farmacologia , Morte Celular , Linhagem Celular Tumoral , Ciclo-Oxigenase 2/metabolismo , Dinoprostona/metabolismo , Relação Dose-Resposta à Radiação , Ativação Enzimática , Fator de Iniciação 4E em Eucariotos/metabolismo , Feminino , Glioma/irrigação sanguínea , Glioma/enzimologia , Glioma/patologia , Células Endoteliais da Veia Umbilical Humana/enzimologia , Humanos , Camundongos Nus , NF-kappa B/metabolismo , Comunicação Parácrina , Fosforilação , Transdução de Sinais/efeitos da radiação , Transfecção , Fator A de Crescimento do Endotélio Vascular/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
18.
Cancer Res ; 76(14): 4124-35, 2016 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-27216177

RESUMO

Cancer-associated fibroblasts (CAF) are components of the tumor microenvironment whose contributions to malignant progression are not fully understood. Here, we show that the fibroblast activation protein (FAP) triggers induction of a CAF subset with an inflammatory phenotype directed by STAT3 activation and inflammation-associated expression signature marked by CCL2 upregulation. Enforcing FAP expression in normal fibroblasts was sufficient to endow them with an inflammatory phenotype similar to FAP(+)CAFs. We identified FAP as a persistent activator of fibroblastic STAT3 through a uPAR-dependent FAK-Src-JAK2 signaling pathway. In a murine liver tumor model, we found that FAP(+)CAFs were a major source of CCL2 and that fibroblastic STAT3-CCL2 signaling in this setting promoted tumor growth by enhancing recruitment of myeloid-derived suppressor cells (MDSC). The CCL2 receptor CCR2 was expressed on circulating MDSCs in tumor-bearing subjects and FAP(+)CAF-mediated tumor promotion and MDSC recruitment was abrogated in Ccr2-deficient mice. Clinically, we observed a positive correlation between stromal expression of FAP, p-STAT3, and CCL2 in human intrahepatic cholangiocarcinoma, a highly aggressive liver cancer with dense desmoplastic stroma, where elevated levels of stromal FAP predicted a poor survival outcome. Taken together, our results showed how FAP-STAT3-CCL2 signaling in CAFs was sufficient to program an inflammatory component of the tumor microenvironment, which may have particular significance in desmoplasia-associated cancers. Cancer Res; 76(14); 4124-35. ©2016 AACR.


Assuntos
Fibroblastos Associados a Câncer/fisiologia , Quimiocina CCL2/fisiologia , Gelatinases/fisiologia , Tolerância Imunológica , Proteínas de Membrana/fisiologia , Fator de Transcrição STAT3/fisiologia , Serina Endopeptidases/fisiologia , Transdução de Sinais/fisiologia , Microambiente Tumoral , Animais , Movimento Celular , Endopeptidases , Feminino , Proteína-Tirosina Quinases de Adesão Focal/fisiologia , Janus Quinase 2/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Receptores de Ativador de Plasminogênio Tipo Uroquinase/fisiologia
19.
Cancer Lett ; 375(1): 31-38, 2016 May 28.
Artigo em Inglês | MEDLINE | ID: mdl-26945967

RESUMO

Pancreatic cancer is a devastating disease characterized by treatment resistance and high recurrence rate. Repopulation of surviving tumor cells undergoing radiotherapy is one of the most common reasons for recurrence. Our previous studies have discovered a novel mechanism for repopulation after irradiation that activation of caspase-3 in irradiated tumor cells activates PKCδ/p38 axis to transmit proliferation signals promoting repopulation of surviving tumor cells. Here we found Sox2 expression is up-regulated in irradiated pancreatic cancer cells, which played a major role in tumor cell repopulation after irradiation. Over-expression of Sox2 strongly enhanced the growth-stimulating effect of irradiated dying tumor cells on living tumor cells through a paracrine modality. Furthermore, we identified activated eIF4E, which is phosphorylated by MNK1, as a regulator of Sox2 expression after irradiation, and pharmacologic inhibition of eIF4E with CGP57380 and Ribavirin significantly weakened Sox2-mediated tumor cell repopulation. Finally, we showed the activation of caspase 3/PKCδ/p38/MNK1 signal pathway in irradiated pancreatic tumor cells. Together, we showed a novel pathway regulating Sox2 expression and Sox2 may be a promising target to reduce recurrence due to repopulation of surviving tumor cells after radiotherapy.


Assuntos
Fator de Iniciação 4E em Eucariotos/metabolismo , Neoplasias Pancreáticas/radioterapia , Fatores de Transcrição SOXB1/metabolismo , Linhagem Celular Tumoral , Proliferação de Células , Técnicas de Cocultura , Regulação Neoplásica da Expressão Gênica/efeitos da radiação , Humanos , Neoplasias Pancreáticas/patologia , Fosforilação , Processamento de Proteína Pós-Traducional , Tolerância a Radiação , Transdução de Sinais , Regulação para Cima
20.
Oncotarget ; 7(13): 16840-54, 2016 Mar 29.
Artigo em Inglês | MEDLINE | ID: mdl-26919238

RESUMO

Thymic stromal lymphopoietin (TSLP) has recently been suggested in several epithelial cancers, either pro-tumor or anti-tumor. However, the role of TSLP in colon cancer remains unknown. We here found significantly decreased TSLP levels in tumor tissues compared with tumor-surrounding tissues of patients with colon cancer and TSLP levels negatively correlated with the clinical staging score of colon cancer. TSLPR, the receptor of TSLP, was expressed in all three colon cancer cell lines investigated and colon tumor tissues. The addition of TSLP significantly enhanced apoptosis of colon cancer cells in a TSLPR-dependent manner. Interestingly, TSLP selectively induced the apoptosis of colon cancer cells, but not normal colonic epithelial cells. Furthermore, we demonstrated that TSLP induced JNK and p38 activation and initiated apoptosis mainly through the extrinsic pathway, as caspase-8 inhibitor significantly reversed the apoptosis-promoting effect of TSLP. Finally, using a xenograft mouse model, we demonstrated that peritumoral administration of TSLP greatly reduced tumor growth accompanied with extensive tumor apoptotic response, which was abolished by tumor cell-specific knockdown of TSLPR. Collectively, our study reveals a novel anti-tumor effect of TSLP via direct promotion of the apoptosis of colon cancer cells, and suggests that TSLP could be of value in treating colon cancer.


Assuntos
Adenoma/patologia , Apoptose/efeitos dos fármacos , Neoplasias do Colo/patologia , Citocinas/farmacologia , Animais , Apoptose/fisiologia , Proliferação de Células/efeitos dos fármacos , Citocinas/metabolismo , Humanos , Camundongos , Camundongos Nus , Receptores de Citocinas/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto , Linfopoietina do Estroma do Timo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA