Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
2.
Nat Cancer ; 5(5): 774-790, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38355776

RESUMO

Pancreatic ductal adenocarcinoma is a highly metastatic disease and macrophages support liver metastases. Efferocytosis, or engulfment of apoptotic cells by macrophages, is an essential process in tissue homeostasis and wound healing, but its role in metastasis is less well understood. Here, we found that the colonization of the hepatic metastatic site is accompanied by low-grade tissue injury and that efferocytosis-mediated clearance of parenchymal dead cells promotes macrophage reprogramming and liver metastasis. Mechanistically, progranulin expression in macrophages is necessary for efficient efferocytosis by controlling lysosomal acidification via cystic fibrosis transmembrane conductance regulator and the degradation of lysosomal cargo, resulting in LXRα/RXRα-mediated macrophage conversion and upregulation of arginase 1. Pharmacological blockade of efferocytosis or macrophage-specific genetic depletion of progranulin impairs macrophage conversion, improves CD8+ T cell functions, and reduces liver metastasis. Our findings reveal how hard-wired functions of macrophages in tissue repair contribute to liver metastasis and identify potential targets for prevention of pancreatic ductal adenocarcinoma liver metastasis.


Assuntos
Carcinoma Ductal Pancreático , Neoplasias Hepáticas , Macrófagos , Neoplasias Pancreáticas , Fagocitose , Microambiente Tumoral , Neoplasias Pancreáticas/patologia , Neoplasias Pancreáticas/metabolismo , Humanos , Neoplasias Hepáticas/secundário , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patologia , Animais , Camundongos , Macrófagos/metabolismo , Carcinoma Ductal Pancreático/patologia , Carcinoma Ductal Pancreático/metabolismo , Linhagem Celular Tumoral , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/metabolismo , Apoptose , Lisossomos/metabolismo , Arginase/metabolismo , Eferocitose
3.
Cancer Res ; 84(4): 527-544, 2024 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-38356443

RESUMO

Pancreatic ductal adenocarcinoma (PDAC) is a highly metastatic disease, yet effective treatments to inhibit PDAC metastasis are lacking. The rich PDAC tumor microenvironment plays a major role in disease progression. Macrophages are the most abundant immune cell population in PDAC tumors and can acquire a range of functions that either hinder or promote tumor growth and metastasis. Here, we identified that mesothelin secretion by pancreatic cancer cells co-opts macrophages to support tumor growth and metastasis of cancer cells to the lungs, liver, and lymph nodes. Mechanistically, secretion of high levels of mesothelin by metastatic cancer cells induced the expression of VEGF alpha (VEGFA) and S100A9 in macrophages. Macrophage-derived VEGFA fed back to cancer cells to support tumor growth, and S100A9 increased neutrophil lung infiltration and formation of neutrophil extracellular traps. These results reveal a role for mesothelin in regulating macrophage functions and interaction with neutrophils to support PDAC metastasis. SIGNIFICANCE: Mesothelin secretion by cancer cells supports pancreatic cancer metastasis by inducing macrophage secretion of VEGFA and S100A9 to support cancer cell proliferation and survival, recruit neutrophils, and stimulate neutrophil extracellular trap formation. See related commentary by Alewine, p. 513.


Assuntos
Carcinoma Ductal Pancreático , Neoplasias Pancreáticas , Humanos , Mesotelina , Linhagem Celular Tumoral , Neoplasias Pancreáticas/patologia , Macrófagos/metabolismo , Carcinoma Ductal Pancreático/patologia , Microambiente Tumoral/fisiologia
4.
Front Cell Dev Biol ; 10: 824075, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35557941

RESUMO

Background: Endometriosis (EMs) is the most common gynaecological disorder with its etiology and/or pathophysiology remains enigmatic. Recent studies showed that extracellular vesicles (EVs), exosomes in particular, play a critical role in developing various clinical disorders. However, the implication of exosomes in endometriosis progression has not been well elucidated. Method: The ectopic stromal cellular exosomes (eEVs) were assessed by transwell assay, scratch tests, tube formation assay, western blot, and qRT-PCR analysis. Protein expression profiles of exosomes in endometrial tissue and vaginal discharge collected from patients with EMS and healthy donors were analysed by Mass spectrometry. siRNA interference technology was used to inhibit the expression of exosomal protein for the functional analysis in in-vivo. Finally, in-vitro experiments were performed to validate the results that we observed in EMs mouse model. Results: In vitro, we discovered that eEVs improved NSC migratory potential by upregulating MMP9 expression and activity. eEVs also aided angiogenesis and elevated the expression of inflammatory cytokines in ovarian epithelial cells, according to our findings. Moesin (MSN) levels in ESC exosomes were substantially greater than in NSC exosomes (1.22e8±5.58e6 vs. 6.605e7±4.574e6, LFQ intensity), as shown by protein mass spectrometry and bioinformatics analysis. In ectopic stromal cells, ERa receptors stimulated the RhoA/Rock-2/MSN pathway. We discovered that downregulating exosomal moesin reduced NSC migration (about 3-fold change) and MMP9 expression (about 2-fold change). On the other hand, Exomsni inhibited angiogenesis and inflammatory cytokine release. In vivo the result of immunohistochemistry and immunofluorescence demonstrated that exosomal MSN substantially modified the expression of MM9, VEGFR and p-VEGFR in polyclonal lesions. In addition, we discovered an elevation in the expression of proinflammatory factors in the surrounding tissue. Conclusion: Exosomal MSN derived from ectopic stromal cells can contribute to endometriosis progression by mediating the construction of a "migration-vascularization-inflammation" loop in the ectopic environment.

5.
J Clin Lab Anal ; 34(11): e23482, 2020 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-32761670

RESUMO

BACKGROUND: Endometriosis (EMS) is a prevalent gynecological condition characterized by the growth of endometrial tissue outside the uterine cavity. This study aimed to clarify the targeted therapeutic effect of sunitinib in an endometriosis in vitro experiment. METHODS: Primary culture of ectopic endometrial cells and normal endometrial cells. Six tumor targeting drugs were selected to screen. MTT was used to determine the IC50, flow cytometry, and DAPI staining of the targeted drugs, in order to determine the apoptosis. The differential proteins after seeding were analyzed by protein spectrum, the correlation between the specific protein and cell apoptosis was determined by small molecule interference, and the expression of each related protein was detected by Western blot. Immunohistochemistry and ELISA were used to detect the expression of p-PDGFR and p-STAT1 in clinical samples, and the correlation between p-STAT1 expression and ectopic focal size was analyzed by SPSS 19. RESULTS: Through the drug screening, it was found that sunitinib has a significant inhibitory effect on ectopic endometrial cells. It was determined that the IC50 of sunitinib on ectopic stromal endometrial cells was 3.32 µM, while the IC50 on normal endometrium was 7.9 µM. Meanwhile, the flow cytometry and DAPI nuclear dye that took out sunitinib had an inhibition effect on the ectopic endometrium at a concentration of 4 µM. Protein spectrum analysis was conducted on ectopic intimal cells after sunitinib treatment, and it was found that STAT1 is specifically expressed in ectopic endometrial cells. In vitro, and through fludarabine interference, it was revealed that sunitinib specifically inhibited the phosphorylation site Tyr751 of PDGFR, while the expression of STAT1, p-STAT1, and caspase-3 was significantly upregulated, and the expression of STAT1 and p-STAT1 was positively correlated with the expression of caspase-3. Finally, the expression of p-PDGFR and p-STAT1 in ectopic foal tissues was both higher than that in normal endometrium, and p-STAT1 expression was positively with ectopic focal size. CONCLUSION: The in vitro experiments revealed that sunitinib could upregulate the expression of STAT1 by inhibiting the phosphorylation site Tyr751 of PDGFR, thereby specifically inducing the apoptosis of the primary heterotopic mesenchymal endometrium.


Assuntos
Apoptose/efeitos dos fármacos , Endométrio , Fator de Transcrição STAT1/metabolismo , Sunitinibe/farmacologia , Biópsia , Células Cultivadas , Endometriose/metabolismo , Endometriose/patologia , Endométrio/citologia , Endométrio/efeitos dos fármacos , Endométrio/metabolismo , Feminino , Humanos , Regulação para Cima/efeitos dos fármacos
6.
Respir Res ; 20(1): 217, 2019 Oct 12.
Artigo em Inglês | MEDLINE | ID: mdl-31606039

RESUMO

BACKGROUND: Icotinib has been widely used in patients with non-small cell lung cancer (NSCLC), and have significantly enhanced the overall survival rate of NSCLC patients. However, acquired drug resistance limits its clinical efficacy. Tumor cell-derived exosomes have been reported to participate in various biological processes, including tumor invasion, metastasis and drug resistance. MATERIALS AND METHODS: In the present study, drug resistance was measured by MTT assay. Exosomes were extracted from the cell supernatant using ultracentrifugation and identified by exosomal marker. HCC827 cells were treated with exosomes derived from icotinib-resistant (IR) HCC827 to observe the invasion and migration of parent cells. The expression of exo-mRNA was analyzed by reverse transcription-quantitative polymerase chain reaction (RT-PCR). In addition, 10 exo-mRNAs detecting from the plasma and bronchoalveolar lavage fluid (BALF) of NSCLC patients with icotinib treatment were used to establish a new drug resistant-warning formula. RESULTS: The oncogene MET into exosomes was identified from icotinib-resistant lung cancer cells, and this was also presented in exosomes in NSCLC patients diagnosed with cancer metastasis after icotinib treatment. The knockdown of MET in exosomes significantly decreased the ability of invasion and migration in HCC827 cells. CONCLUSION: It was suggested that MET might be specifically package and transferred by exosomes to modify the invasion and migration ability of the surrounding icotinib-sensitive cells.


Assuntos
Antineoplásicos Imunológicos/farmacologia , Movimento Celular/genética , Éteres de Coroa/farmacologia , Resistencia a Medicamentos Antineoplásicos/genética , Exossomos/metabolismo , Invasividade Neoplásica/genética , Oncogenes/genética , Proteínas Proto-Oncogênicas c-met/biossíntese , Proteínas Proto-Oncogênicas c-met/genética , Quinazolinas/farmacologia , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Líquido da Lavagem Broncoalveolar/química , Linhagem Celular Tumoral , Ensaios de Seleção de Medicamentos Antitumorais , Regulação Neoplásica da Expressão Gênica , Humanos , Neoplasias Pulmonares/patologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA