Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 370
Filtrar
1.
Microbiol Spectr ; 9(3): e0149721, 2021 12 22.
Artigo em Inglês | MEDLINE | ID: mdl-34937192

RESUMO

Tumors and infectious agents both benefit from an immunosuppressive environment. Cutibacterium acnes (C. acnes) is a bacterium in the normal skin microbiota, which has the ability to survive intracellularly in macrophages and is significantly more common in prostate cancer tissue compared with normal prostate tissue. This study investigated if prostate cancer tissue culture positive for C. acnes has a higher infiltration of regulatory T-cells (Tregs) and if macrophages stimulated with C. acnes induced the expression of immunosuppressive genes that could be linked to an increase of Tregs in prostate cancer. Real-time PCR and enzyme-linked immunosorbent spot assay (ELISA) were used to examine the expression of immunosuppressive genes in human macrophages stimulated in vitro with C. acnes, and associations between the presence of C. acnes and infiltration of Tregs were investigated by statistically analyzing data generated in two previous studies. The in vitro results demonstrated that macrophages stimulated with C. acnes significantly increased their expression of PD-L1, CCL17, and CCL18 mRNA and protein (p <0.05). In the cohort, Tregs in tumor stroma and tumor epithelia were positively associated with the presence of C. acnes (P = 0.0004 and P = 0.046, respectively). Since the macrophages stimulated with C. acnes in vitro increased the expression of immunosuppressive genes, and prostate cancer patients with prostatic C. acnes infection had higher infiltration of Tregs than their noninfected counterparts, we suggest that C. acnes may contribute to an immunosuppressive tumor environment that is vital for prostate cancer progression. IMPORTANCE In an immune suppressive tumor microenvironment constituted by immunosuppressive cells and immunosuppressive mediators, tumors may improve their ability to give rise to a clinically relevant cancer. In the present study, we found that C. acnes might contribute to an immunosuppressive environment by recruiting Tregs and by increasing the expression of immunosuppressive mediators such as PD-L1, CCL17, and CCL18. We believe that our data add support to the hypothesis of a contributing role of C. acnes in prostate cancer development. If established that C. acnes stimulates prostate cancer progression it may open up avenues for targeted prostate cancer treatment.


Assuntos
Tolerância Imunológica/imunologia , Macrófagos/imunologia , Propionibacteriaceae/imunologia , Neoplasias da Próstata/imunologia , Neoplasias da Próstata/microbiologia , Linfócitos T Reguladores/imunologia , Antígeno B7-H1/biossíntese , Antígeno B7-H1/genética , Quimiocina CCL17/biossíntese , Quimiocina CCL17/genética , Quimiocinas CC/biossíntese , Quimiocinas CC/genética , ELISPOT , Humanos , Tolerância Imunológica/genética , Masculino , Microbiota/imunologia , Neoplasias da Próstata/patologia , Evasão Tumoral/imunologia , Microambiente Tumoral/imunologia
2.
Cell Death Dis ; 10(11): 796, 2019 10 22.
Artigo em Inglês | MEDLINE | ID: mdl-31641099

RESUMO

CCL14 is a member of CC chemokines and its role in hepatocellular carcinoma (HCC) is still unknown. In this study, CCL14 expression were analyzed by tissue microarray (TMA) including 171 paired tumor and peritumor tissues of patients from Zhongshan Hospital of Fudan University. We found for the first time that CCL14 was downregulated in HCC tumor tissues compared with peritumor tissues (P = 0.01). Meanwhile, CCL14 low expression in HCC tumor tissues is associated with a poor prognosis (P = 0.035). CCL14 also displayed its predictive value in high differentiation (P = 0.026), liver cirrhosis (P = 0.003), and no tumor capsule (P = 0.024) subgroups. The underlying mechanisms were further investigated in HCC cell lines by CCL14 overexpression and knock-down in vitro. We found overexpression of CCL14 suppressed proliferation and promoted apoptosis of HCC cells. Finally, the effect was confirmed by animal xenograft tumor models in vivo. The results shown overexpression of CCL14 lead to inhibiting the growth of tumor in nude mice. Interestingly, our data also implied that CCL14 played these effects by inhibiting the activation of Wnt/ß-catenin pathway. These findings suggest CCL14 is a novel prognostic factor of HCC and serve as a tumor suppressor.


Assuntos
Carcinoma Hepatocelular/metabolismo , Quimiocinas CC/metabolismo , Neoplasias Hepáticas/metabolismo , Animais , Apoptose/fisiologia , Biomarcadores Tumorais/biossíntese , Biomarcadores Tumorais/genética , Biomarcadores Tumorais/metabolismo , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/patologia , Ciclo Celular/fisiologia , Linhagem Celular Tumoral , Quimiocinas CC/biossíntese , Quimiocinas CC/genética , Regulação para Baixo , Feminino , Células Hep G2 , Xenoenxertos , Humanos , Imuno-Histoquímica , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/patologia , Masculino , Camundongos , Camundongos Nus , Prognóstico
3.
Cancer Chemother Pharmacol ; 82(1): 99-109, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29728799

RESUMO

PURPOSE: High activity of enzyme TOP2a in tumor cells is known to be associated with sensitivity to anthracycline chemotherapy, but 20% of such patients do not show clinical response. Tumor microenvironment, including tumor-associated macrophages (TAM), is an essential factor defining the efficiency of chemotherapy. In the present study, we analyzed the expression of M2 macrophage markers, YKL-39 and CCL18, in tumors of breast cancer patients received anthracycline-based NAC. METHODS: Patients were divided into two groups according to the level of doxorubicin sensitivity marker TOP2a: DOX-Sense and DOX-Res groups. Expression levels of TOR2a, CD68, YKL-39 and CCL18 genes were analyzed by qPCR, the amplification of TOR2a gene locus was assessed by the microarray assay. Clinical and pathological responses to neoadjuvant chemotherapy were assessed. RESULTS: We found that the average level of TOP2a expression in patients of DOX-Sense group was almost 10 times higher than in patients of DOX-Res group, and the expression of CD68 was 3 times higher in the DOX-Sense group compared to DOX-Res group. We demonstrated that expression levels of M2-derived cytokines but not the amount of TAM is indicative for clinical and pathological chemotherapy efficacy in breast cancer patients. Out of 8 patients from DOX-Sense group who did not respond to neoadjuvant chemotherapy (NAC), 7 patients had M2+ macrophage phenotype (YKL-39+CCL18- or YKL-39-CCL18+) and only one patient had M2- macrophage phenotype (YKL-39-CCL18-). In DOX-Res group, out of 14 patients who clinically responded to NAC 9 patients had M2- phenotype and only 5 patients had M2+ macrophage phenotype. Among pathological non-responders in DOX-Sense group, 19 (82%) patients had M2+ tumor phenotype and only 4 (18%) patients had M2- phenotype. In DOX-Res group, all 5 patients who pathologically responded to NAC had M2 phenotype (YKL-39-CCL18-). Unlike the clinical response to NAC, the differences in the frequency of M2+ and M2- phenotypes between pathologically responding and non-responding patients within DOX-Sense and DOX-Res groups were statistically significant. CONCLUSIONS: Thus, we showed that in patients with breast cancer who received anthracycline-containing NAC the absence of clinical response is associated with the presence of M2+ macrophage phenotype (YKL-39-CCL18 + or YKL-39 + CCL18-) based on TOP2a overexpression data.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/metabolismo , Quimiocinas CC/biossíntese , Quitinases/biossíntese , Macrófagos/metabolismo , Neoplasias da Mama/genética , Neoplasias da Mama/imunologia , Capecitabina/administração & dosagem , Polaridade Celular/imunologia , Quimiocinas CC/genética , Quimiocinas CC/imunologia , Quimioterapia Adjuvante , Quitinases/genética , Quitinases/imunologia , Ciclofosfamida/administração & dosagem , DNA Topoisomerases Tipo II/biossíntese , DNA Topoisomerases Tipo II/genética , Doxorrubicina/administração & dosagem , Feminino , Fluoruracila/administração & dosagem , Amplificação de Genes , Expressão Gênica , Humanos , Ativação de Macrófagos , Macrófagos/imunologia , Macrófagos/patologia , Pessoa de Meia-Idade , Terapia Neoadjuvante , Estadiamento de Neoplasias , Proteínas de Ligação a Poli-ADP-Ribose/biossíntese , Proteínas de Ligação a Poli-ADP-Ribose/genética
4.
Cancer Biomark ; 21(4): 925-934, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29504526

RESUMO

BACKGROUND: Molecular target therapy has become a hot spot in cancer treatment, finding effective targets for diffuse large B cell lymphoma (DLBCL) is an urgent problem. OBJECTIVE: To detect the expression level of C-C motif chemokine ligand 18 (CCL18) in DLBCL and clarify its potential role in the progression of DLBCL. METHODS: Gene expression datas of DLBCL were obtained from TCGA and GEO databases. The relationship between CCL18 and clinicopathologic information of DLBCL was assessed using meta-analysis method. Then we conducted bioinformatics analysis to uncover the biological function of CCL18 and its co-expression genes. Immunohistochemistry was applied to detect expression of CCL18 in DLBCL and reactive hyperplasia lymphoid tissues. RESULTS: The expression of CCL18 in DLBCL was higher than negative control group. The levels of CCL18 were distinct in different molecular subtypes and ages, and patients with higher level of CCL18 had a shorter overall survival than those with lower level. CCL18 and its co-expression genes were enriched in biological function such as cell proliferation, migration, apoptotic, and correlated with NF-κB, pathway in cancer, PI3K-AKT pathway. CONCLUSIONS: CCL18 was up-regulated in DLBCL and related to poor prognosis. CCL18 may act as a valuable target for diagnosis and treatment of DLBCL.


Assuntos
Biomarcadores Tumorais/análise , Quimiocinas CC/biossíntese , Linfoma Difuso de Grandes Células B/patologia , Adulto , Idoso , Progressão da Doença , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Prognóstico , Regulação para Cima
5.
Tumour Biol ; 39(7): 1010428317708900, 2017 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-28718365

RESUMO

Hepatocellular carcinoma is one of the most mortal and prevalent cancers with increasing incidence worldwide. Elucidating genetic driver genes for prognosis and palindromia of hepatocellular carcinoma helps managing clinical decisions for patients. In this study, the high-throughput RNA sequencing data on platform IlluminaHiSeq of hepatocellular carcinoma were downloaded from The Cancer Genome Atlas with 330 primary hepatocellular carcinoma patient samples. Stable key genes with differential expressions were identified with which Kaplan-Meier survival analysis was performed using Cox proportional hazards test in R language. Driver genes influencing the prognosis of this disease were determined using clustering analysis. Functional analysis of driver genes was performed by literature search and Gene Set Enrichment Analysis. Finally, the selected driver genes were verified using external dataset GSE40873. A total of 5781 stable key genes were identified, including 156 genes definitely related to prognoses of hepatocellular carcinoma. Based on the significant key genes, samples were grouped into five clusters which were further integrated into high- and low-risk classes based on clinical features. TMEM88, CCL14, and CLEC3B were selected as driver genes which clustered high-/low-risk patients successfully (generally, p = 0.0005124445). Finally, survival analysis of the high-/low-risk samples from external database illustrated significant difference with p value 0.0198. In conclusion, TMEM88, CCL14, and CLEC3B genes were stable and available in predicting the survival and palindromia time of hepatocellular carcinoma. These genes could function as potential prognostic genes contributing to improve patients' outcomes and survival.


Assuntos
Biomarcadores Tumorais/genética , Carcinoma Hepatocelular/genética , Quimiocinas CC/genética , Lectinas Tipo C/genética , Neoplasias Hepáticas/genética , Proteínas de Membrana/genética , Biomarcadores Tumorais/biossíntese , Carcinoma Hepatocelular/patologia , Quimiocinas CC/biossíntese , Intervalo Livre de Doença , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Estimativa de Kaplan-Meier , Lectinas Tipo C/biossíntese , Neoplasias Hepáticas/patologia , Masculino , Proteínas de Membrana/biossíntese , Prognóstico , Modelos de Riscos Proporcionais
6.
Artigo em Inglês | MEDLINE | ID: mdl-28553619

RESUMO

CCL23, also known as myeloid progenitor inhibitory factor (MPIF)-1, macrophage inflammatory protein (MIP)-3, or CKß8, is a member of the CC chemokine subfamily exerting its effects via CCR1 binding. By doing so, CCL23 selectively recruits resting T lymphocytes and monocytes, inhibits proliferation of myeloid progenitor cells and promotes angiogenesis. Previously, we and other groups have reported that human neutrophils are able to produce chemokines upon appropriate activation, including CCR1-binding CCL2, CCL3, and CCL4. Herein, we demonstrate that human neutrophils display the capacity to also express and release CCL23 when stimulated by R848 and, to a lesser extent, by other pro-inflammatory agonists, including LPS, Pam3CSK4, and TNFα. Notably, we show that, on a per cell basis, R848-activated neutrophils produce higher levels of CCL23 than autologous CD14+-monocytes activated under similar experimental conditions. By contrast, we found that, unlike CD14+-monocytes, neutrophils do not produce CCL23 in response to IL-4, thus indicating that they express CCL23 in a stimulus-specific fashion. Finally, we show that the production of CCL23 by R848-stimulated neutrophils is negatively modulated by IFNα, which instead enhances that of CCL2. Together, data extend our knowledge on the chemokines potentially produced by neutrophils. The ability of human neutrophils to produce CCL23 further supports the notion on the neutrophil capacity of orchestrating the recruitment of different cell types to the inflamed sites, in turn contributing to the control of the immune response.


Assuntos
Quimiocinas CC/biossíntese , Imidazóis/farmacologia , Neutrófilos/imunologia , Neutrófilos/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Células Cultivadas , Quimiocinas/metabolismo , Quimiocinas CC/genética , Quimiocinas CC/metabolismo , Humanos , Interleucina-4/metabolismo , Cinética , Lipopeptídeos/metabolismo , Lipopolissacarídeos/metabolismo , Neutrófilos/efeitos dos fármacos , RNA Mensageiro/biossíntese , Receptores CCR1 , Transdução de Sinais/imunologia , Receptores Toll-Like/agonistas
7.
J Immunol ; 198(10): 4115-4128, 2017 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-28396316

RESUMO

Decidual spiral arteriole (SpA) remodeling is essential to ensure optimal uteroplacental blood flow during human pregnancy, yet very little is known about the regulatory mechanisms. Uterine decidual NK (dNK) cells and macrophages infiltrate the SpAs and are proposed to initiate remodeling before colonization by extravillous trophoblasts (EVTs); however, the trigger for their infiltration is unknown. Using human first trimester placenta, decidua, primary dNK cells, and macrophages, we tested the hypothesis that EVTs activate SpA endothelial cells to secrete chemokines that have the potential to recruit maternal immune cells into SpAs. Gene array, real-time PCR, and ELISA analyses showed that treatment of endothelial cells with EVT conditioned medium significantly increased production of two chemokines, CCL14 and CXCL6. CCL14 induced chemotaxis of both dNK cells and decidual macrophages, whereas CXCL6 also induced dNK cell migration. Analysis of the decidua basalis from early pregnancy demonstrated expression of CCL14 and CXCL6 by endothelial cells in remodeling SpAs, and their cognate receptors are present in both dNK cells and macrophages. Neutralization studies identified IL-6 and CXCL8 as factors secreted by EVTs that induce endothelial cell CCL14 and CXCL6 expression. This study has identified intricate crosstalk between EVTs, SpA cells, and decidual immune cells that governs their recruitment to SpAs in the early stages of remodeling and has identified potential key candidate factors involved. This provides a new understanding of the interactions between maternal and fetal cells during early placentation and highlights novel avenues for research to understand defective SpA remodeling and consequent pregnancy pathology.


Assuntos
Arteríolas/fisiologia , Decídua/fisiologia , Células Endoteliais/metabolismo , Células Matadoras Naturais/fisiologia , Macrófagos/fisiologia , Trofoblastos/metabolismo , Arteríolas/citologia , Arteríolas/imunologia , Movimento Celular/imunologia , Células Cultivadas , Quimiocina CXCL6/biossíntese , Quimiocina CXCL6/imunologia , Quimiocinas CC/biossíntese , Quimiocinas CC/imunologia , Meios de Cultura/química , Decídua/imunologia , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/imunologia , Feminino , Humanos , Interleucina-6/imunologia , Interleucina-6/metabolismo , Interleucina-8/imunologia , Interleucina-8/metabolismo , Macrófagos/imunologia , Placenta/citologia , Placenta/imunologia , Gravidez , Primeiro Trimestre da Gravidez , Trofoblastos/imunologia
8.
Biomed Mater ; 12(2): 025006, 2017 03 07.
Artigo em Inglês | MEDLINE | ID: mdl-28267684

RESUMO

After implantation of a biomaterial, an inflammatory response involving macrophages is induced. The behavior of macrophages depends on their phenotype, and by directing macrophage polarization unwanted effects may be avoided. In this study, the possibility to modulate the behavior of macrophages activated by biomaterials was assessed in an in vitro model. Primary human monocytes were seeded on polyethylene terephthalate, polypropylene and polylactic acid yarns, and treated with medications frequently used by patients: rapamycin, dexamethasone, celecoxib or pravastatin. Modulation of the adhering macrophages with rapamycin resulted in a generally pro-inflammatory effect. Dexamethasone caused an overall anti-inflammatory effect on the macrophages cultured on either material, while celecoxib only affected macrophages adhering to polyethylene terephthalate and polylactic acid. Pravastatin increased the pro-inflammatory genes of macrophages cultured on polypropylene and polylactic acid. Pairwise comparison revealed that macrophages adhering to polylactic acid seemed to be more susceptible to phenotype modulation than when adhering to polypropylene or polyethylene terephthalate. The data show that macrophages activated by the biomaterials can be modulated, yet the degree of the modulatory capacity depends on the type of material. Combined, this model provides insights into the possibility of using a medication in combination with a biomaterial to direct macrophage behavior and thereby possibly avoid unwanted effects after implantation.


Assuntos
Materiais Biocompatíveis/química , Macrófagos/efeitos dos fármacos , Macrófagos/fisiologia , Celecoxib/farmacologia , Adesão Celular/genética , Adesão Celular/fisiologia , Células Cultivadas , Quimiocinas CC/biossíntese , Quimiocinas CC/genética , Dexametasona/farmacologia , Expressão Gênica/efeitos dos fármacos , Humanos , Mediadores da Inflamação/fisiologia , Interleucina-6/biossíntese , Interleucina-6/genética , Ativação de Macrófagos/efeitos dos fármacos , Ativação de Macrófagos/fisiologia , Teste de Materiais , Monócitos/efeitos dos fármacos , Monócitos/fisiologia , Fenótipo , Poliésteres/química , Polietilenotereftalatos/química , Polipropilenos/química , Pravastatina/farmacologia , Sirolimo/farmacologia
9.
Clin Cancer Res ; 23(3): 833-844, 2017 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-27492974

RESUMO

PURPOSE: We have reported loss of SMAD4 promotes expression of CCL15 from colorectal cancer to recruit CCR1+ myeloid cells through the CCL15-CCR1 axis, which contributes to invasion and liver metastasis. However, the molecular mechanism of lung metastasis is yet to be elucidated. Our purpose is to determine whether similar mechanism is involved in the lung metastasis of colorectal cancer. EXPERIMENTAL DESIGN: In a mouse model, we examined whether SMAD4 could affect the metastatic activity of colorectal cancer cells to the lung through the CCL15-CCR1 axis. We immunohistochemically analyzed expression of SMAD4, CCL15, and CCR1 with 107 clinical specimens of colorectal cancer lung metastases. We also characterized the CCR1+ myeloid cells using several cell-type-specific markers. RESULTS: In a mouse model, CCL15 secreted from SMAD4-deficient colorectal cancer cells recruited CCR1+ cells, promoting their metastatic activities to the lung. Immunohistochemical analysis of lung metastases from colorectal cancer patients revealed that CCL15 expression was significantly correlated with loss of SMAD4, and that CCL15-positive metastases recruited approximately 1.9 times more numbers of CCR1+ cells than CCL15-negative metastases. Importantly, patients with CCL15-positive metastases showed a significantly shorter relapse-free survival (RFS) than those with CCL15-negative metastases, and multivariate analysis indicated that CCL15 expression was an independent predictor of shorter RFS. Immunofluorescent staining showed that most CCR1+ cells around lung metastases were tumor-associated neutrophil, although a minor fraction was granulocytic myeloid-derived suppressor cell. CONCLUSIONS: CCL15-CCR1 axis may be a therapeutic target to prevent colorectal cancer lung metastasis. CCL15 can be a biomarker indicating poor prognosis of colorectal cancer patients with lung metastases. Clin Cancer Res; 23(3); 833-44. ©2016 AACR.


Assuntos
Quimiocinas CC/fisiologia , Neoplasias Colorretais/patologia , Neoplasias Pulmonares/secundário , Proteínas Inflamatórias de Macrófagos/fisiologia , Proteínas de Neoplasias/deficiência , Infiltração de Neutrófilos , Receptores CCR1/fisiologia , Proteína Smad4/deficiência , Animais , Linhagem Celular Tumoral , Movimento Celular , Quimiocinas CC/biossíntese , Quimiocinas CC/genética , Neoplasias Colorretais/metabolismo , Intervalo Livre de Doença , Feminino , Regulação Neoplásica da Expressão Gênica , Genes Reporter , Xenoenxertos , Humanos , Neoplasias Pulmonares/imunologia , Neoplasias Pulmonares/metabolismo , Proteínas Inflamatórias de Macrófagos/biossíntese , Proteínas Inflamatórias de Macrófagos/genética , Camundongos , Camundongos Nus , Camundongos SCID , Células Mieloides/metabolismo , Proteínas de Neoplasias/biossíntese , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/fisiologia , Prognóstico , Modelos de Riscos Proporcionais , Proteína Smad4/fisiologia
10.
Int J Mol Sci ; 17(11)2016 Nov 12.
Artigo em Inglês | MEDLINE | ID: mdl-27845745

RESUMO

Allergic asthma is an inflammatory lung disease that is partly sustained by the chemokine eotaxin-3 (CCL26), which extends eosinophil migration into tissues long after allergen exposure. Modulation of CCL26 could represent a means to mitigate airway inflammation. Here we evaluated procyanidin A2 as a means of modulating CCL26 production and investigated interactions with the known inflammation modulator, Interferon γ (IFNγ). We used the human lung epithelial cell line A549 and optimized the conditions for inducing CCL26. Cells were exposed to a range of procyanidin A2 or IFNγ concentrations for varied lengths of time prior to an inflammatory insult of interleukin-4 (IL-4) for 24 h. An enzyme-linked immunosorbent assay was used to measure CCL26 production. Exposing cells to 5 µM procyanidin A2 (prior to IL-4) reduced CCL26 production by 35% compared with control. Greatest inhibition by procyanidin A2 was seen with a 2 h exposure prior to IL-4, whereas IFNγ inhibition was greatest at 24 h. Concomitant incubation of procyanidin A2 and IFNγ did not extend the inhibitory efficacy of procyanidin A2. These data provide evidence that procyanidin A2 can modulate IL-4-induced CCL26 production by A549 lung epithelial cells and that it does so in a manner that is different from IFNγ.


Assuntos
Catequina/farmacologia , Quimiocinas CC/biossíntese , Fatores Imunológicos/farmacologia , Interleucina-4/fisiologia , Proantocianidinas/farmacologia , Células A549 , Asma/tratamento farmacológico , Asma/imunologia , Quimiocina CCL26 , Quimiocinas CC/genética , Avaliação Pré-Clínica de Medicamentos , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Expressão Gênica , Humanos , Alvéolos Pulmonares/citologia
11.
Arch Oral Biol ; 66: 77-85, 2016 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-26921718

RESUMO

OBJECTIVE: P. gingivalis is a gram-negative anaerobic bacterium and a major periodontal pathogen. LPS produced by P. gingivalis promotes osteoclast formation. TECK is a CC chemokine whose expression is increased in gingival epithelial cells exposed to P. gingivalis LPS. In this study, we investigated the effect of TECK in osteoclastogenesis induced by P. gingivalis LPS. DESIGNS: Real time reverse transcriptase polymerase chain reaction (RTPCR) analysis and western blotting were performed to confirm TECK in MG63, human osteoblast cell line and primary murine osteoblasts and CCR9 in RAW 264.7 cells and murine bone marrow macrophages (BMMs) as osteoclast precursors. P. gingivalis LPS-treated BMMs and Raw 264.7 cells were cultured with or without TECK or TECK antibody to examine the effect of TECK on osteoclast formation. Cocultures with murine osteoblasts and bone marrow cells were also treated with or without TECK or TECK antibody. Luciferase assay and western blotting were used to determine whether TECK-CCR9 induced osteoclastogenesis was mediated through NFATc1 or NF-kB signaling. RESULTS: TECK was shown to be expressed by osteoblasts, and its receptor, CCR9, by osteoclast precursors. TECK increased P. gingivalis LPS-induced osteoclast numbers in an in vitro osteoclast formation assay using osteoclast precursors. The enhanced osteoclast formation by TECK was mediated by NFATc1, but not by NF-kB signaling. CONCLUSION: TECK may be a novel regulator of osteoclast formation induced by P. gingivalis LPS in periodontitis.


Assuntos
Quimiocinas CC/farmacologia , Lipopolissacarídeos/farmacologia , Fatores de Transcrição NFATC/metabolismo , Osteoclastos/efeitos dos fármacos , Porphyromonas gingivalis/fisiologia , Animais , Células da Medula Óssea/efeitos dos fármacos , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular , Quimiocinas CC/biossíntese , Gengiva/citologia , Gengiva/metabolismo , Humanos , Macrófagos/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos BALB C , NF-kappa B/metabolismo , Osteoblastos/citologia , Osteoblastos/efeitos dos fármacos , Osteoblastos/metabolismo , Osteoclastos/citologia , Osteoclastos/metabolismo , Osteoclastos/microbiologia , Osteogênese , Porphyromonas gingivalis/efeitos dos fármacos , Células RAW 264.7 , Receptores CCR/biossíntese , Proteínas Recombinantes/farmacologia , Transdução de Sinais/efeitos dos fármacos
12.
BMC Cancer ; 16: 98, 2016 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-26875556

RESUMO

BACKGROUND: The differential diagnosis of follicular thyroid carcinoma (FTC) and follicular adenoma (FA) before surgery is a clinical challenge. Many efforts have been made but most focusing on tumor cells, while the roles of tumor associated macrophages (TAMs) remained unclear in FTC. Here we analyzed the differences between TAMs in FTC and those in FA. METHODS: We first analyzed the density of TAMs by CD68 immunostaining in 59 histologically confirmed FTCs and 47 FAs. Cytokines produced by FTC and FA were profiled using antibody array, and validated by quantitative PCR. Chemotaxis of monocyte THP-1 was induced by condition medium of FTC cell lines (FTC133 and WRO82-1) with and without anti-CCL15 neutralizing antibody. Finally, we analyzed CCL15 protein level in FTC and FA by immunohistochemistry. RESULTS: The average density of CD68(+) cells was 9.5 ± 5.4/field in FTC, significantly higher than that in FA (4.9 ± 3.4/field, p < 0.001). Subsequently profiling showed that CCL15 was the most abundant chemokine in FTC compared with FA. CCL15 mRNA in FTC was 51.4-folds of that in FA. CM of FTC cell lines induced THP-1 cell chemotaxis by 33 ~ 77%, and anti-CCL15 neutralizing antibody reduced THP-1 cell migration in a dose-dependent manner. Moreover, we observed positive CCL15 immunostaining in 67.8% of FTCs compared with 23.4% of FAs. CONCLUSION: Our study suggested FTC might induce TAMs infiltration by producing CCL15. Measurement of TAMs and CCL15 in follicular thyroid lesions may be applied clinically to differentiate FTC from FA pre-operation.


Assuntos
Adenocarcinoma Folicular/diagnóstico , Adenoma/diagnóstico , Quimiocinas CC/biossíntese , Diagnóstico Diferencial , Proteínas Inflamatórias de Macrófagos/biossíntese , Adenocarcinoma Folicular/genética , Adenocarcinoma Folicular/patologia , Adenoma/genética , Adenoma/patologia , Biópsia por Agulha Fina , Quimiocinas CC/genética , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Proteínas Inflamatórias de Macrófagos/genética , Macrófagos/patologia , Masculino , Período Pré-Operatório , RNA Mensageiro/biossíntese , Análise Serial de Tecidos
13.
Stem Cells ; 34(4): 1112-22, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26763650

RESUMO

Mesenchymal stem cells (MSCs) possess the ability to migrate toward tumor sites and are regarded as promising gene delivery vehicles for cancer therapeutics. However, the factors that mediate this tropism have yet to be completely elucidated. In this study, through cytokine array analysis, chemokine CCL15 was found to be the most abundant protein differentially expressed in hepatocellular carcinoma (HCC) cell lines compared with a normal liver cell line. Serum CCL15 levels in HCC patients determined by enzyme linked immunosorbent assay were shown to be profoundly elevated compared with healthy controls. Immunohistochemical analysis indicated that CCL15 expression was much stronger in HCC tumor tissues than in adjacent nontumor tissues. Transwell migration assay suggested that CCL15 may be involved in chemotaxis of human MSCs (hMSCs) toward HCC in vitro and that this chemotactic effect of CCL15 is mediated via CCR1 receptors on hMSCs. Orthotopic animal models of HCC were established to investigate the role of CCL15 in hMSCs migration toward HCC in vivo. Both histological and flow cytometric analysis showed that significantly fewer hMSCs localized within 97H-CCL15-shRNA xenografts compared with 97H-green fluorescent protein xenografts after intravenous delivery. Finally, the possible effects of hMSCs on HCC tumor growth were also evaluated. Coculture experiments showed that hMSCs had no apparent effect on the proliferation of HCC cells in vitro In addition, systemic administration of hMSCs did not affect HCC tumor progression in vivo. Our data in this study help to elucidate the mechanism underlying the homing capacity of hMSCs toward HCC.


Assuntos
Carcinoma Hepatocelular/terapia , Quimiocinas CC/genética , Técnicas de Transferência de Genes , Neoplasias Hepáticas/terapia , Proteínas Inflamatórias de Macrófagos/genética , Células-Tronco Mesenquimais/metabolismo , Animais , Células da Medula Óssea/citologia , Células da Medula Óssea/metabolismo , Carcinoma Hepatocelular/genética , Linhagem Celular Tumoral , Movimento Celular/genética , Quimiocinas CC/biossíntese , Quimiocinas CC/uso terapêutico , Quimiotaxia/genética , Regulação Neoplásica da Expressão Gênica , Proteínas de Fluorescência Verde/genética , Humanos , Neoplasias Hepáticas/genética , Proteínas Inflamatórias de Macrófagos/biossíntese , Proteínas Inflamatórias de Macrófagos/uso terapêutico , Células-Tronco Mesenquimais/química , Células-Tronco Mesenquimais/citologia , Camundongos , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/uso terapêutico , Receptores CCR1/biossíntese , Receptores CCR1/genética , Ensaios Antitumorais Modelo de Xenoenxerto
14.
Protein Expr Purif ; 119: 1-10, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26506568

RESUMO

In the continued absence of an effective anti-HIV vaccine, approximately 2 million new HIV infections occur every year, with over 95% of these in developing countries. Calls have been made for the development of anti-HIV drugs that can be formulated for topical use to prevent HIV transmission during sexual intercourse. Because these drugs are principally destined for use in low-resource regions, achieving production costs that are as low as possible is an absolute requirement. 5P12-RANTES, an analog of the human chemokine protein RANTES/CCL5, is a highly potent HIV entry inhibitor which acts by achieving potent blockade of the principal HIV coreceptor, CCR5. Here we describe the development and optimization of a scalable low-cost production process for 5P12-RANTES based on expression in Pichia pastoris. At pilot (150 L) scale, this cGMP compliant process yielded 30 g of clinical grade 5P12-RANTES. As well as providing sufficient material for the first stage of clinical development, this process represents an important step towards achieving production of 5P12-RANTES at a cost and scale appropriate to meet needs for topical HIV prevention worldwide.


Assuntos
Fármacos Anti-HIV/metabolismo , Quimiocinas CC/biossíntese , Infecções por HIV/tratamento farmacológico , HIV/efeitos dos fármacos , Pichia , Fármacos Anti-HIV/isolamento & purificação , Fármacos Anti-HIV/farmacologia , Reatores Biológicos/economia , Reatores Biológicos/normas , Quimiocinas CC/isolamento & purificação , Quimiocinas CC/farmacologia , Cromatografia Líquida de Alta Pressão , Cromatografia por Troca Iônica , Fermentação , Humanos , Concentração Inibidora 50 , Projetos Piloto , Internalização do Vírus/efeitos dos fármacos
15.
Cancer Res ; 75(24): 5283-98, 2015 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-26483204

RESUMO

Tumor cell survival in the hostile distant organ is a rate-limiting step in cancer metastasis. Bone marrow-derived myeloid cells can form a premetastatic niche and provide a tumor-promoting microenvironment. However, it is unclear whether these myeloid cells in the premetastatic site have any direct effect on tumor cell survival. Here, we report that chemokine CCL9 was highly induced in Gr-1(+)CD11b(+) immature myeloid cells and in premetastatic lung in tumor-bearing mice. Knockdown of CCL9 in myeloid cells decreased tumor cell survival and metastasis. Importantly, CCL9 overexpression in myeloid cells lacking TGFß signaling rescued the tumor metastasis defect observed in mice with myeloid-specific Tgfbr2 deletion. The expression level of CCL23, the human orthologue for CCL9, in peripheral blood mononuclear cells correlated with progression and survival of cancer patients. Our study demonstrates that CCL9 could serve as a good candidate for anti-metastasis treatment by targeting the rate-limiting step of cancer cell survival. In addition, targeting CCL9 may avoid the adverse effects of TGFß-targeted therapy.


Assuntos
Quimiocinas CC/metabolismo , Proteínas Inflamatórias de Macrófagos/metabolismo , Células Mieloides/metabolismo , Invasividade Neoplásica/patologia , Neoplasias Experimentais/patologia , Fator de Crescimento Transformador beta/metabolismo , Animais , Western Blotting , Linhagem Celular Tumoral , Sobrevivência Celular , Quimiocinas CC/biossíntese , Modelos Animais de Doenças , Ensaio de Imunoadsorção Enzimática , Citometria de Fluxo , Imunofluorescência , Humanos , Marcação In Situ das Extremidades Cortadas , Camundongos , Invasividade Neoplásica/imunologia , Neoplasias Experimentais/imunologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais/fisiologia , Transfecção , Microambiente Tumoral/imunologia
16.
Pathol Biol (Paris) ; 63(4-5): 199-203, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26294068

RESUMO

PURPOSE OF THE STUDY: To investigate whether CCL18 is involved in breast cancer, and the relationship between CCL18 and MVD (MVD was recognized by CD34) which is a well-accepted angiogenic maker of multiple cancers including breast cancer. PATIENTS AND METHODS: Immunohistochemistry staining for CCL18 and CD34 was performed on 179 cases, including 29 normal cases as control, 47 cases with benign breast diseases, and 103 cases with breast cancer. RESULTS: We found that CCL18 was significantly up-regulated in breast cancer samples as compared with benign tumors or normal breast tissues. Moreover, the expression level of CCL18 increased with the size of tumors, the number of lymph node metastasis, and advancing tumor stage, suggesting that CCL18 expression correlates with tumor malignancy scales. At the same time, we found that MVD was also significantly over-expressed in cancer tissues as compared with normal control group and benign tumor group, but it was not significantly differentially expressed among tumors with different malignancy scale like CCL18, while the expression of MVD in CCL18 positive breast cancer cases was higher than in the CCL18 negative breast cancer cases (P=0.016, P<0.05). CONCLUSION: CCL18 is involved in the development of breast cancer. CCL18 is a better biomarker than MVD in determining whether the tumor is malignant and the severity of malignancy of breast cancer.


Assuntos
Neoplasias da Mama/metabolismo , Carcinoma/metabolismo , Quimiocinas CC/fisiologia , Proteínas de Neoplasias/fisiologia , Adulto , Antígenos CD34/análise , Biomarcadores Tumorais , Mama/metabolismo , Doenças Mamárias/embriologia , Neoplasias da Mama/irrigação sanguínea , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Carcinoma/irrigação sanguínea , Carcinoma/genética , Carcinoma/patologia , Quimiocinas CC/biossíntese , Quimiocinas CC/genética , Progressão da Doença , Feminino , Humanos , Metástase Linfática , Pessoa de Meia-Idade , Proteínas de Neoplasias/biossíntese , Proteínas de Neoplasias/genética , Estadiamento de Neoplasias , Neovascularização Patológica/patologia , Carga Tumoral
17.
Mediators Inflamm ; 2015: 176926, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26300589

RESUMO

Asthma is associated with increased pulmonary inflammation and airway hyperresponsiveness. The interaction between airway epithelium and inflammatory mediators plays a key role in the pathogenesis of asthma. In vitro studies evaluated the inhibitory effects of 3-(2,5-dimethoxyphenyl)-1-(5-methylfuran-2-yl)prop-2-en-1-one (DMPF-1), a synthetic chalcone analogue, upon inflammation in the A549 lung epithelial cell line. DMPF-1 selectively inhibited TNF-α-stimulated CC chemokine secretion (RANTES, eotaxin-1, and MCP-1) without any effect upon CXC chemokine (GRO-α and IL-8) secretion. Western blot analysis further demonstrated that the inhibitory activity resulted from disruption of p65NF-κB nuclear translocation without any effects on the mitogen-activated protein kinase (MAPK) pathway. Treatment of ovalbumin-sensitized and ovalbumin-challenged BALB/c mice with DMPF-1 (0.2-100 mg/kg) demonstrated significant reduction in the secretion and gene expression of CC chemokines (RANTES, eotaxin-1, and MCP-1) and Th2 cytokines (IL-4, IL-5, and IL-13). Furthermore, DMPF-1 treatment inhibited eosinophilia, goblet cell hyperplasia, peripheral blood total IgE, and airway hyperresponsiveness in ovalbumin-sensitized and ovalbumin-challenged mice. In conclusion, these findings demonstrate the potential of DMPF-1, a nonsteroidal compound, as an antiasthmatic agent for further pharmacological evaluation.


Assuntos
Asma/tratamento farmacológico , Núcleo Celular/metabolismo , Chalconas/farmacologia , Quimiocinas CC/antagonistas & inibidores , Furanos/farmacologia , Cetonas/farmacologia , NF-kappa B/metabolismo , Transporte Ativo do Núcleo Celular/efeitos dos fármacos , Animais , Hiper-Reatividade Brônquica/prevenção & controle , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Quimiocinas CC/biossíntese , Eosinófilos/fisiologia , Feminino , Humanos , Imunoglobulina E/sangue , Pulmão/imunologia , Pulmão/patologia , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos BALB C
18.
Am J Respir Cell Mol Biol ; 53(5): 676-88, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25870903

RESUMO

Macrophages are dynamic cells that mature under the influence of signals from the local microenvironment into either classically (M1) or alternatively (M2) activated macrophages with specific functional and phenotypic properties. Although the phenotypic identification of M1 and M2 macrophages is well established in mice, this is less clear for human macrophages. In addition, the persistence and reversibility of polarized human phenotypes is not well established. Human peripheral blood monocytes were differentiated into uncommitted macrophages (M0) and then polarized to M1 and M2 phenotypes using LPS/IFN-γ and IL-4/IL-13, respectively. M1 and M2 were identified as CD64(+)CD80(+) and CD11b(+)CD209(+), respectively, by flow cytometry. Polarized M1 cells secreted IP-10, IFN-γ, IL-8, TNF-α, IL-1ß, and RANTES, whereas M2 cells secreted IL-13, CCL17, and CCL18. Functionally, M2 cells were highly endocytic. In cytokine-deficient medium, the polarized macrophages reverted back to the M0 state within 12 days. If previously polarized macrophages were given the alternative polarizing stimulus after 6 days of resting in cytokine-deficient medium, a switch in polarization was seen (i.e., M1 macrophages switched to M2 and expressed CD11b(+)CD209(+) and vice versa). In summary, we report phenotypic identification of human M1 and M2 macrophages, their functional characteristics, and their ability to be reprogrammed given the appropriate stimuli.


Assuntos
Ativação de Macrófagos/efeitos dos fármacos , Macrófagos/efeitos dos fármacos , Monócitos/efeitos dos fármacos , Diferenciação Celular , Quimiocina CCL17/biossíntese , Quimiocina CCL17/metabolismo , Quimiocina CCL5/biossíntese , Quimiocina CCL5/metabolismo , Quimiocina CXCL10/biossíntese , Quimiocina CXCL10/metabolismo , Quimiocinas CC/biossíntese , Quimiocinas CC/metabolismo , Endocitose/efeitos dos fármacos , Endocitose/imunologia , Expressão Gênica/efeitos dos fármacos , Expressão Gênica/imunologia , Humanos , Imunofenotipagem , Interferon gama/farmacologia , Interleucina-13/farmacologia , Interleucina-1beta/biossíntese , Interleucina-1beta/metabolismo , Interleucina-4/farmacologia , Interleucina-8/biossíntese , Interleucina-8/metabolismo , Lipopolissacarídeos/farmacologia , Macrófagos/citologia , Macrófagos/imunologia , Monócitos/citologia , Monócitos/imunologia , Cultura Primária de Células , Fator de Necrose Tumoral alfa/biossíntese , Fator de Necrose Tumoral alfa/metabolismo
19.
J Med Virol ; 87(5): 790-7, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25678365

RESUMO

The human T-cell lymphotropic virus type 1 (HTLV-1) and HTLV-2 are common copathogens among Human Immunodeficiency Virus (HIV)-infected individuals. HTLV-2 may confer a survival benefit among patients with HIV-1/HTLV-2 coinfections, along with lower plasma HIV-1 levels and delayed rates of CD4(+) T-cell decline. These effects have been attributed to the ability of the HTLV-2 viral transactivating Tax2 protein to induce the production of high levels of antiviral CC-chemokines and to downregulate expression of the CCR5 receptor, resulting in impaired entry of HIV-1 into CD4(+) T-cells. This study investigated the innate immunity of coinfected HIV/HTLV individuals by testing the ability of patient PBMCs to produce CC-chemokines in association CCR5 receptor modulation. The cellular proliferative responses of HIV/HTLV coinfected versus HIV monoinfected individuals were also evaluated. Higher levels of MIP-1α, MIP-1ß, and RANTES (P < 0.05) were found in HIV-1/HTLV-2 coinfected group compared to HIV-1 monoinfected population. Upregulated levels of RANTES were shown in HIV-1/HTLV-1 after 1 and 3 days of culture (P < 0.05). Lymphocytes from HIV-1/HTLV-2 coinfected individuals showed significant CCR5 downregulation after 1 and 3 days of culture compared to lymphocytes from HIV-1 and uninfected groups (P < 0.05). Lower percentages of CCR5-positive cells were found in HIV-1/HTLV-1 coinfected after 3 days of incubation (P < 0.05). Levels of proliferation were significantly higher in the HIV-1/HTLV-1 group compared to HIV-1 alone (P < 0.05). HTLV-2 and HTLV-1 infections may induce the involvement of innate immunity against HIV-1 via stimulation of CC-chemokines and receptors, potentially modifying CCR5/HIV-1 binding and HIV-1 progression in coinfected individuals.


Assuntos
Quimiocinas CC/biossíntese , Coinfecção/imunologia , Infecções por HIV/imunologia , Infecções por HTLV-I/imunologia , Infecções por HTLV-II/imunologia , Receptores CCR5/biossíntese , Adulto , Idoso , Proliferação de Células , Coinfecção/virologia , Feminino , Perfilação da Expressão Gênica , Infecções por HIV/complicações , Infecções por HIV/virologia , HIV-1/isolamento & purificação , Infecções por HTLV-I/complicações , Infecções por HTLV-I/virologia , Infecções por HTLV-II/complicações , Infecções por HTLV-II/virologia , Vírus Linfotrópico T Tipo 1 Humano/isolamento & purificação , Vírus Linfotrópico T Tipo 2 Humano/isolamento & purificação , Humanos , Leucócitos Mononucleares/imunologia , Masculino , Pessoa de Meia-Idade
20.
Allergy ; 70(4): 374-83, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25564944

RESUMO

BACKGROUND: Asthma is characterized by chronic airway inflammation and airway hyperresponsiveness (AHR). Little is known about the role of pulmonary stem/progenitor cells (PSCs) in allergic airway inflammation. METHODS: To identify and investigate the role of PSCs in the bronchial epithelium of neonatal mice, we developed an enzyme-based digestion method to obtain single-cell suspension from lung tissues. Characterization of PSCs was performed using flow cytometry, real-time PCR, immunofluorescence staining, confocal microscopy, and scanning electron microscopy. The effects of SSEA-1(+) (stage-specific embryonic antigen-1) PSCs was studied in an in vivo model of ovalbumin-induced allergic inflammation and an in vitro model of cell-based regulation using flow cytometry, real-time PCR, and immune-blotting. RESULTS: Single-cell suspensions derived from neonatal lung tissue included populations that expressed either SSEA-1(+) or Sca-1(+) (stem cell antigen-1). The SSEA-1(+) PSCs were highly prevalent in neonatal mice, and they were rare in adult mice. Enriched neonatal SSEA-1(+) PSCs had the ability of self-renewal and differentiated into pneumocytes and tracheal epithelial cells. SSEA-1(+) PSCs reduced AHR and airway damage in asthmatic mice by decreasing eosinophil infiltration, inhibiting chemokines/cytokines production, and preserving the level of CCSP. CONCLUSIONS: Here, we demonstrated that neonatal SSEA-1(+) PSCs play an immunomodulatory role in the progression of asthma by reducing lung damage and inhibiting inflammatory responses. Further understanding the molecular mechanisms of neonatal SSEA-1(+) PSCs might shed light on exploring the novel therapeutic approaches for allergic airway inflammation.


Assuntos
Antígenos CD15/metabolismo , Pulmão/citologia , Pulmão/metabolismo , Hipersensibilidade Respiratória/imunologia , Hipersensibilidade Respiratória/metabolismo , Células-Tronco/metabolismo , Animais , Animais Recém-Nascidos , Autorrenovação Celular , Quimiocinas CC/biossíntese , Evolução Clonal , Citocinas/biossíntese , Modelos Animais de Doenças , Imunofenotipagem , Camundongos , Células-Tronco Multipotentes/citologia , Células-Tronco Multipotentes/metabolismo , Fenótipo , Hipersensibilidade Respiratória/diagnóstico , Hipersensibilidade Respiratória/terapia , Mucosa Respiratória/citologia , Mucosa Respiratória/imunologia , Mucosa Respiratória/metabolismo , Índice de Gravidade de Doença , Transplante de Células-Tronco , Células-Tronco/citologia , Linfopoietina do Estroma do Timo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA