Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Exp Hematol Oncol ; 13(1): 48, 2024 May 09.
Artigo em Inglês | MEDLINE | ID: mdl-38725070

RESUMO

BACKGROUND: Cancer is the leading cause of death among older adults. Although the integration of immunotherapy has revolutionized the therapeutic landscape of cancer, the complex interactions between age and immunotherapy efficacy remain incompletely defined. Here, we aimed to elucidate the relationship between aging and immunotherapy resistance. METHODS: Flow cytometry was performed to evaluate the infiltration of immune cells in the tumor microenvironment (TME). In vivo T cell proliferation, cytotoxicity and migration assays were performed to evaluate the antitumor capacity of tumor antigen-specific CD8+ T cells in mice. Real-time quantitative PCR (qPCR) was used to investigate the expression of IFN-γ-associated gene and natural killer (NK)-associated chemokine. Adoptive NK cell transfer was adopted to evaluate the effects of NK cells from young mice in overcoming the immunotherapy resistance of aged mice. RESULTS: We found that elderly patients with advanced non-small cell lung cancer (aNSCLC) aged ≥ 75 years exhibited poorer progression-free survival (PFS), overall survival (OS) and a lower clinical response rate after immunotherapy. Mechanistically, we showed that the infiltration of NK cells was significantly reduced in aged mice compared to younger mice. Furthermore, the aged NK cells could also suppress the activation of tumor antigen-specific CD8+ T cells by inhibiting the recruitment and activation of CD103+ dendritic cells (DCs). Adoptive transfer of NK cells from young mice to aged mice promoted TME remodeling, and reversed immunotherapy resistance. CONCLUSION: Our findings revealed the decreased sensitivity of elderly patients to immunotherapy, as well as in aged mice. This may be attributed to the reduction of NK cells in aged mice, which inhibits CD103+ DCs recruitment and its CD86 expression and ultimately leads to immunotherapy resistance.

2.
Clin Proteomics ; 21(1): 35, 2024 May 19.
Artigo em Inglês | MEDLINE | ID: mdl-38764042

RESUMO

BACKGROUND: Currently, no effective measures are available to predict the curative efficacy of small-cell lung cancer (SCLC) chemotherapy. We expect to develop a method for effectively predicting the SCLC chemotherapy efficacy and prognosis in clinical practice in order to offer more pertinent therapeutic protocols for individual patients. METHODS: We adopted matrix-assisted laser desorption/ionization time-of-flight mass spectrometry (MALDI-TOF-MS) and ClinPro Tools system to detect serum samples from 154 SCLC patients with different curative efficacy of standard chemotherapy and analyze the different peptides/proteins of SCLC patients to discover predictive tumor markers related to chemotherapy efficacy. Ten peptide/protein peaks were significantly different in the two groups. RESULTS: A genetic algorithm model consisting of four peptides/proteins was developed from the training group to separate patients with different chemotherapy efficacies. Among them, three peptides/proteins (m/z 3323.35, 6649.03 and 6451.08) showed high expression in the disease progression group, whereas the peptide/protein at m/z 4283.18 was highly expressed in the disease response group. The classifier exhibited an accuracy of 91.4% (53/58) in the validation group. The survival analysis showed that the median progression-free survival (PFS) of 30 SCLC patients in disease response group was 9.0 months; in 28 cases in disease progression group, the median PFS was 3.0 months, a statistically significant difference (χ2 = 46.98, P < 0.001). The median overall survival (OS) of the two groups was 13.0 months and 7.0 months, a statistically significant difference (χ2 = 40.64, P < 0.001). CONCLUSIONS: These peptides/proteins may be used as potential biological markers for prediction of the curative efficacy and prognosis for SCLC patients treated with standard regimen chemotherapy.

3.
J Environ Manage ; 360: 121153, 2024 May 20.
Artigo em Inglês | MEDLINE | ID: mdl-38772234

RESUMO

Strategic coordination between urbanization and carbon emission efficiency (CEE) is vital for promoting low-carbon urbanization and sustainable urban planning. In order to assess the coupled coordination degree (CCD) of urbanization and CEE and investigate the factors influencing the CCD, this research employs the Super slacks-based measure (SBM) model, the coupled coordination degree model (CCDM), and the Tobit model. Four key findings emerge from the analysis of the temporal and spatial evolution traits of the CCD based on data from 106 nations worldwide between 2005 and 2020. (1) The global CEE shows a significant downward trend, and the spatial disparity is unambiguous. high CEE countries hang in the north and west of Europe, while those in Asia, Africa and the east of Europe have lower CEE. (2) The combined urbanization level and demographic, economic and social urbanization are all on an upward trend. Singapore has the greatest degree of urbanization overall globally. (3) The CCD of urbanization and CEE shows a fluctuating upward trend, with particularly strong changes in 2018-2020. 2017 and 2018 are the years with better global coupling coordination status. During the study period, the CCD results of countries are mostly uncoordinated and low coordination, and the CCD of the United States, China, India and Japan is in the front. (4) The effect of urban electrification rate on the CCD is positive; the effect of foreign trade and net inflow of foreign direct investment is negative; while energy structure and industrial structure have no significant effect. A number of policy proposals are put forth in light of the outcomes of the research to enhance the coordination.

4.
Front Immunol ; 15: 1371559, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38576625

RESUMO

Anti-PD-(L)1 therapy has shown great efficacy in some patients with cancer. However, a significant proportion of patients with cancer do not respond to it. Another unmet clinical need for anti-PD-(L)1 therapy is the dynamic monitoring of treatment effects. Therefore, identifying biomarkers that can stratify potential responders before PD-(L)1 treatment and timely monitoring of the efficacy of PD-(L)1 treatment are crucial in the clinical setting. The identification of biomarkers by liquid biopsy has attracted considerable attention. Among the identified biomarkers, circulating T cells are one of the most promising because of their indispensable contribution to anti-PD-(L)1 therapy. The present review aimed to thoroughly explore the potential of circulating T cells as biomarkers of anti-PD-(L)1 therapy and its advantages and limitations.


Assuntos
Neoplasias , Linfócitos T , Humanos , Biomarcadores , Imunoterapia , Neoplasias/tratamento farmacológico
5.
Sci Transl Med ; 15(679): eabn5029, 2023 01 18.
Artigo em Inglês | MEDLINE | ID: mdl-36652534

RESUMO

Anti-PD-1/PD-L1 therapy, either by anti-PD-1 antibody or anti-PD-L1 antibody, has efficacy by reinvigorating tumor-infiltrating CD8+ T cells in a subset of patients with cancer, but it has unequal effects on heterogeneous CD8+ T cell populations. Hence, the subset crucial to efficacious PD-1 blockade therapy remains elusive. Here, we found an increase in tumor-infiltrating CD200+ cytotoxic T lymphocytes (CTLs) upon PD-1/PD-L1 blockade, with higher proportions of CD200+ T cells positively related to a favorable clinical outcome to anti-PD-1/PD-L1 therapy in three independent cohorts of patients with cancer. Using multiple mouse tumor models, we demonstrated that CD200+ CTLs are essential for efficacious anti-PD-L1 therapy. Mechanistically, we observed a unique chromatin landscape in CD200+ CTLs and found that these cells are enriched for tumor antigen-specific CTLs and have antitumor effector functions. Coinoculation of CD200+ CTLs with tumor cells led to robust tumor regression in two transplanted mouse models. Clinically, we found that infiltration of CD200+ CTLs into tumors could predict immunotherapy efficacy in six patient cohorts. Together, our findings reveal that CD200+ CTLs in the tumor microenvironment are crucial for efficacious anti-PD-1/PD-L1 therapy and could serve as a predictor of successful immunotherapy in the clinic.


Assuntos
Neoplasias , Linfócitos T Citotóxicos , Animais , Camundongos , Linfócitos T CD8-Positivos , Microambiente Tumoral , Neoplasias/terapia , Imunoterapia , Antígeno B7-H1 , Linfócitos do Interstício Tumoral
6.
RSC Adv ; 13(1): 615-623, 2022 Dec 19.
Artigo em Inglês | MEDLINE | ID: mdl-36605644

RESUMO

Fabrication of a dense polymer/ceramic composite membrane with high permeability remains a great challenge. In this study, a highly selective polydopamine (PDA)/ceramic composite nanofiltration (NF) membrane was prepared by using an Al2O3 ceramic membrane with pore size of 0.1 µm as the support layer. In order to improve the membrane formation rate, KMnO4 was introduced to oxidize the dopamine to improve the reactivity, and Na2CO3 was used to adjust the pH value of the dopamine solution. When the addition amount of KMnO4 is 0.2 g L-1 and that of Na2CO3 is 1 g L-1, a functional layer can be formed within 10 min. PDA and polyethyleneimine (PEI) were added to the functional layer to adjust the selectivity of the composite membrane. The composite membrane showed a rejection of 99.7% towards Congo red dye with a high flux of 165 L (m2 h bar)-1 at ambient temperature. After 3 h treatment with Congo red, the fouling resistance of the membrane was improved compared with that of the ceramic based membrane. The surface morphology and composition of the composite membrane were also characterized with scanning electron microscopy (SEM) and X-ray photoelectron spectroscopy (XPS), which confirmed the successful preparation of the PDA/ceramic composite membrane.

7.
Front Oncol ; 11: 585271, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33718140

RESUMO

The efficacy of current treatment regimens for pancreatic cancer (PC) remains unsatisfactory. In recent years, immune checkpoint blockade (ICB) therapy has shown promising anti-tumor outcomes in many malignancies, including PC. Inexpensive and readily available biomarkers which predict therapeutic responses and prognosis are in critical need. Systemic immune-inflammation index (SII) and neutrophil-lymphocyte ratio (NLR) are emerging predictors for prognosis of various tumors. We aim to investigate the prognostic significance of baseline SII, NLR, and their changes in PC patients treated with ICB. Our retrospective analysis included PC patients treated with ICB therapy in the Chinese PLA General Hospital. All demographic, biological, and clinical data were extracted from medical records. Relative changes of SII after two doses of ICB were defined as ΔSII% and calculated as (SIIafter 2 doses-SIIbaseline)/SIIbaseline, and so was the case for ΔNLR%. Overall survival (OS) and progression-free survival (PFS) were compared using Kaplan-Meier curves. The prognostic significance of baseline SII, NLR, and their changes was assessed in univariate and multivariate analyses using the Cox proportional hazard regression model. In total, 122 patients with PC treated with ICB were included in the present analysis. Elevated baseline SII (HR=3.28; 95% CI:1.98-5.27; P=0.03) and ΔNLR% (HR=2.21; 95% CI:1.03-4.74; P=0.04) were significantly correlated with an increased risk of death. For PC patients receiving ICB combined with chemotherapies or radiotherapies as the first-line treatment, increased baseline SII was a negative predictor for both OS (HR=8.06; 95% CI:1.71-37.86; P=0.01) and PFS (HR=2.84; 95%CI:1.37-10.38; P=0.04). Our study reveals the prognostic value of baseline SII and NLR changes in PC patients receiving ICB therapy. The clinical utility of these prognostic biomarkers needs to be further studied in prospective studies.

8.
J Immunother Cancer ; 9(2)2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33593825

RESUMO

Programmed cell death 1 (PD-1)-based immunotherapy has revolutionized the treatment of various cancers. However, only a certain group of patients benefit from PD-1 blockade therapy and many patients succumb to hyperprogressive disease. Although, CD8 T cells and conventional T cells are generally considered to be the primary source of PD-1 in cancer, accumulating evidence suggests that other distinct cell types, including B cells, regulatory T cells, natural killer cells, dendritic cells, tumor-associated macrophages and cancer cells, also express PD-1. Hence, the response of patients with cancer to PD-1 blockade therapy is a cumulative effect of anti-PD-1 antibodies acting on a myriad of cell types. Although, the contribution of CD8 T cells to PD-1 blockade therapy has been well-established, recent studies also suggest the involvement of non-canonical PD-1 signaling in blockade therapy. This review discusses the role of non-canonical PD-1 signaling in distinct cell types and explores how the available knowledge can improve PD-1 blockade immunotherapy, particularly in identifying novel biomarkers and combination treatment strategies.


Assuntos
Inibidores de Checkpoint Imunológico/uso terapêutico , Neoplasias/metabolismo , Transdução de Sinais/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Inibidores de Checkpoint Imunológico/farmacologia , Neoplasias/tratamento farmacológico , Receptor de Morte Celular Programada 1/metabolismo , Microambiente Tumoral/efeitos dos fármacos
9.
Front Immunol ; 11: 582657, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33193391

RESUMO

In immune system, B cells are classically positive modulators that regulate inflammation and immune responses. Regulatory B cells (Bregs) are a subset of B cells which play crucial roles in various conditions, including infection, allergies, autoimmune diseases, transplantation, and tumors. Until now, unequivocal surface markers for Bregs still lack consensus, although numerous Breg subsets have been identified. Generally, Bregs exert their immunoregulatory functions mainly through cytokine secretion and intercellular contact. In the tumor microenvironment, Bregs suppress effector T cells, induce regulatory T cells and target other tumor-infiltrating immune cells, such as myeloid-derived suppressor cells, natural killer cells and macrophages, to hamper anti-tumor immunity. Meanwhile, the cross-regulations between Bregs and tumor cells often result in tumor escape from immunosurveillance. In addition, accumulating evidence suggests that Bregs are closely associated with many clinicopathological factors of cancer patients and might be potential biomarkers for accessing patient survival. Thus, Bregs are potential therapeutic targets for future immunotherapy in cancer patients. In this review, we will discuss the phenotypes, functions, and clinical relevance of Bregs in cancer.


Assuntos
Linfócitos B Reguladores/imunologia , Neoplasias/imunologia , Linfócitos T Reguladores/imunologia , Animais , Biomarcadores Tumorais , Humanos , Tolerância Imunológica , Ativação Linfocitária , Neoplasias/diagnóstico , Fenótipo , Microambiente Tumoral
10.
J Exp Clin Cancer Res ; 39(1): 11, 2020 Jan 13.
Artigo em Inglês | MEDLINE | ID: mdl-31931851

RESUMO

BACKGROUND: Mounting evidence suggests that complement components promote tumor progression via modulating immune suppression, angiogenesis, or tumor cell proliferation. However, the role of C3a-C3aR signaling in regulating lung metastasis of breast cancer remains unknown. METHODS: We performed various ex-vivo and in-vivo assays. Genetic and pharmacological C3aR blockade models were applied to investigate the role of C3a-C3aR in metastasis of breast cancer. RESULTS: C3a-C3aR signaling in CAFs facilitates the metastasis of breast cancer. Mechanically, C3a-C3aR signaling augments pro-metastatic cytokine secretion and extracellular matrix components expression of CAFs via the activation of PI3K-AKT signaling. Genetic or pharmacological blockade of C3aR signaling effectively inhibited lung metastasis of breast cancer in mouse models. CONCLUSIONS: C3a-C3aR signaling in CAFs facilitates the metastasis of breast cancer. Targeting C3aR signaling is a potential anti-metastasis strategy for breast cancer therapy.


Assuntos
Neoplasias da Mama/patologia , Fibroblastos Associados a Câncer/metabolismo , Complemento C3/metabolismo , Neoplasias Pulmonares/patologia , Neoplasias Pulmonares/secundário , Receptores de Complemento/metabolismo , Animais , Arginina/administração & dosagem , Arginina/análogos & derivados , Arginina/farmacologia , Compostos Benzidrílicos/administração & dosagem , Compostos Benzidrílicos/farmacologia , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/genética , Neoplasias da Mama/metabolismo , Fibroblastos Associados a Câncer/efeitos dos fármacos , Linhagem Celular Tumoral , Complemento C3/genética , Citocinas/genética , Citocinas/metabolismo , Proteínas da Matriz Extracelular/genética , Proteínas da Matriz Extracelular/metabolismo , Feminino , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/metabolismo , Camundongos , Invasividade Neoplásica , Transplante de Neoplasias , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Receptores de Complemento/genética , Transdução de Sinais
11.
Cancer Immunol Res ; 7(2): 193-207, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30514794

RESUMO

Complement aids in the construction of an immunosuppressive tumor microenvironment. Tumor cell-derived C3 has been previously reported, but whether and how it acts on antitumor immunity remains to be elucidated. Here, we describe a mechanism for tumor cell-derived C3 in suppressing antitumor immunity. Tumor cell-derived C3 was activated intracellularly, which results in generation of C3a. C3a modulated tumor-associated macrophages via C3a-C3aR-PI3Kγ signaling, thereby repressing antitumor immunity. Deletion of C3 in tumor cells that had high C3 expression enhanced efficacy of anti-PD-L1 treatment. Collectively, our results suggest tumor cell-derived C3 may be a useful target for cancer immunotherapy and that targeting C3 in tumor cells may enhance antitumor immunity.


Assuntos
Antineoplásicos Imunológicos/farmacologia , Antígeno B7-H1/antagonistas & inibidores , Ativação do Complemento/imunologia , Complemento C3/imunologia , Macrófagos/imunologia , Neoplasias/imunologia , Animais , Antígeno B7-H1/metabolismo , Classe Ib de Fosfatidilinositol 3-Quinase/metabolismo , Modelos Animais de Doenças , Feminino , Humanos , Imunomodulação , Imunoterapia , Macrófagos/metabolismo , Melanoma Experimental , Camundongos , Neoplasias/metabolismo , Neoplasias/patologia , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/metabolismo , Subpopulações de Linfócitos T/patologia , Microambiente Tumoral/imunologia
12.
Cancer Manag Res ; 10: 6971-6984, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30588102

RESUMO

PURPOSE: Centromere protein U (CENPU) abnormally exhibits high expression in various types of human tumor tissues and participates in tumor progression; however, its expression pattern and biological function in lung cancer have not yet been elucidated. In the present study, we explored the clinical significance and biological function of CENPU in lung cancer. MATERIALS AND METHODS: The Cancer Genome Atlas (TCGA) data analyses, quantitative real-time PCR (RT-PCR), and Western blotting were performed to quantify CENPU and FOXM1 expression in non-small-cell lung cancer (NSCLC) samples. Survival data were obtained from Kaplan-Meier plotter or PROGgene V2 prognostic database. The function of CENPU in lung cancer cell proliferation was determined using 5-ethynyl-2'-deoxyuridine (EdU), Cell Counting Kit-8 (CCK-8), and cell cycle assays, and the underlying mechanism was determined through bioinformatic analyses and validated by in vitro siRNA or plasmid transfection experiments. RESULTS: CENPU was abnormally overexpressed in NSCLC samples compared with matched paired normal tissues. Higher expression of CENPU predicted worse overall survival (OS) and relapse-free survival (RFS) in NSCLC patients. Knockdown of CENPU expression by siRNA significantly inhibited proliferation and delayed cell cycle progression of lung cancer cells. To figure out the mechanism, bioinformatic analyses were performed and the results showed that the transcription factor, FOXM1, positively correlated with CENPU. Further in vitro experiments indicated that FOXM1 was the possible downstream transcription factor of CENPU as the knockdown of CENPU led to lower expression of FOXM1 and the overexpression of FOXM1 significantly reversed the inhibition of proliferation caused by CENPU knockdown. Furthermore, FOXM1 was highly expressed in NSCLC. The knockdown of FOXM1 also attenuated proliferation and induced G1 arrest in lung cancer cells. CONCLUSION: CENPU was highly expressed in NSCLC tissues, wherein it promoted lung cancer cell proliferation via the transcription factor, FOXM1, which could be a potential target for therapeutic strategies.

13.
Front Immunol ; 9: 87, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29441068

RESUMO

Fibrinogen-like protein 2 (Fgl2) is critical for immune regulation in the inflammatory state. Elevated Fgl2 levels are observed in patients with inflammatory bowel disease (IBD), but little is known about its functional significance. In this study, we sought to investigate the role of Fgl2 in the development of intestinal inflammation and colitis-associated colorectal cancer (CAC). Here, we report that Fgl2 deficiency increased susceptibility to dextran sodium sulfate-induced colitis and CAC in a mouse model. During colitis development, the expression of the membrane-bound and secreted forms of Fgl2 (mFgl2 and sFgl2, respectively) in the colon were increased and predominantly expressed by colonic macrophages. In addition, using bone marrow chimeric mice, we determined that Fgl2 function in colitis is strictly related to its expression in the hematopoietic cells. Loss of Fgl2 induced the polarization of M1, but suppressed that of M2 both in vivo and in vitro, independent of intestinal inflammation. Thus, Fgl2 suppresses intestinal inflammation and CAC development through its role in macrophage polarization and may serve as a therapeutic target in inflammatory diseases, including IBD.


Assuntos
Transformação Celular Neoplásica/imunologia , Transformação Celular Neoplásica/metabolismo , Colite/imunologia , Colite/metabolismo , Fibrinogênio/genética , Macrófagos/imunologia , Macrófagos/metabolismo , Animais , Biomarcadores , Células da Medula Óssea/imunologia , Células da Medula Óssea/metabolismo , Colite/induzido quimicamente , Colite/complicações , Sulfato de Dextrana/efeitos adversos , Modelos Animais de Doenças , Expressão Gênica , Humanos , Mucosa Intestinal/imunologia , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patologia , Ativação de Macrófagos/genética , Ativação de Macrófagos/imunologia , Camundongos , Camundongos Knockout
14.
Oncoimmunology ; 6(10): e1349587, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-29123963

RESUMO

Anti-PD-1/PD-L1 therapy has achieved great success in the clinic; however, only a small fraction of cancer patient benefit from PD-1/PD-L1 blockade therapy, and overcoming resistance to PD-1/PD-L1 blockade has thus become a primary priority. In this study, we demonstrated that administration of PD-1/PD-L1 antibodies resulted in the activation of the complement system and massive generation of C5a. Generation of C5a did not change the accumulation of MDSCs in either the tumor or spleen but enhanced their inhibitory potential. In addition, blockade of C5a-C5aR signaling in combination with PD-1/PD-L1 antibodies greatly enhanced the anti-tumor efficacy of PD-1/PD-L1 antibodies. Overall, these data indicate an immunosuppressive role of C5a in the context of PD-1/PD-L1 blockade therapy and provide a strong incentive to clinically explore combination therapies using a C5a antagonist.

15.
Int J Biol Sci ; 13(6): 804-814, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28656005

RESUMO

Fibrinogen-like protein 2 (Fgl2), a member of the fibrinogen super family, is a pleiotropic cytokine that impacts diverse cellular functions. Previous studies have shown that tumor cell-derived Fgl2 promotes tumorigenesis and metastasis in immune-deficient mice, and it also functions as an immune-suppressive modulator in glioblastoma multiform (GMB). This study aimed to evaluate whether and how tumor stroma-derived Fgl2 affects tumorigenesis and tumor progression. We established the syngeneic transplantable Lewis lung carcinoma (LLC) model in Fgl2-knock-out (Fgl2-KO) mice and we found that deficiency of host Fgl2 is associated with reduced growth of syngeneic LLC tumors. Furthermore, we confirmed that host Fgl2 deficiency significantly decreased the accumulation of myeloid-derived suppressor cells (MDSCs) through down-regulation of chemokine (C-X-C motif) ligand 12 (CXCL12) expression. More importantly, we demonstrated that Fgl2 induced an activated and pro-tumorigenic phenotype of cancer-associated fibroblasts (CAFs) which are the principal source of CXCL12 in the tumor microenvironment (TME). Our results present a novel role of stroma-derived Fgl2 in CAF activation and function, suggesting that Fgl2 is an effective therapeutic target for treating lung cancer.


Assuntos
Fibroblastos Associados a Câncer/metabolismo , Fibroblastos Associados a Câncer/patologia , Fibrinogênio/metabolismo , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patologia , Animais , Apoptose/genética , Apoptose/fisiologia , Western Blotting , Carcinoma Pulmonar de Lewis/metabolismo , Linhagem Celular , Quimiocina CXCL12/metabolismo , Feminino , Fibrinogênio/genética , Citometria de Fluxo , Masculino , Camundongos , Camundongos Knockout , Células Supressoras Mieloides/metabolismo , Microambiente Tumoral
16.
Int J Mol Sci ; 18(1)2017 Jan 19.
Artigo em Inglês | MEDLINE | ID: mdl-28106852

RESUMO

The initiator of extrinsic coagulation, tissue factor (TF), and its non-coagulant isoform alternatively spliced TF (asTF) are closely associated with tumor development. In the tumor microenvironment, the role of TF-induced coagulation in tumor progression remains to be fully elucidated. Using TF-knockdown lung tumor cells, we showed that TF is the dominant component of procoagulant activity but is dispensable in the cellular biology of tumor cells. In a xenograft model, using immunohistochemical analysis and flow cytometry analysis of the tumor microenvironment, we demonstrated that TF-induced fibrin deposition, which is correlated with complement activation and myeloid-derived suppressor cell (MDSC) recruitment, is positively associated with tumor progression. C5aR antagonism blunted the effect of TF on tumor progression and decreased MDSC recruitment. In conclusion, our data suggested that in tumor microenvironment, TF-induced coagulation activated the complement system and subsequently recruited myeloid-derived suppressor cells to promote tumor growth, which brings new insights into the coagulation-induced complement activation within the tumor microenvironment during tumor progression.


Assuntos
Ativação do Complemento , Progressão da Doença , Neoplasias Pulmonares/sangue , Neoplasias Pulmonares/patologia , Células Supressoras Mieloides/metabolismo , Tromboplastina/metabolismo , Células A549 , Animais , Apoptose , Coagulação Sanguínea , Proliferação de Células , Feminino , Técnicas de Silenciamento de Genes , Humanos , Camundongos Nus , Receptor da Anafilatoxina C5a/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...