Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 41
Filtrer
1.
mBio ; 14(5): e0164523, 2023 Oct 31.
Article de Anglais | MEDLINE | ID: mdl-37772878

RÉSUMÉ

IMPORTANCE: Sliding clamp is a highly conserved protein in the evolution of prokaryotic and eukaryotic cells. The sliding clamp is required for genomic replication as a critical co-factor of DNA polymerases. However, the sliding clamp analogs in viruses remain largely unknown. We found that the ASFV E301R protein (pE301R) exhibited a sliding clamp-like structure and similar functions during ASFV replication. Interestingly, pE301R is assembled into a unique ring-shaped homotetramer distinct from sliding clamps or proliferating cell nuclear antigens (PCNAs) from other species. Notably, the E301R gene is required for viral life cycle, but the pE301R function can be partially restored by the porcine PCNA. This study not only highlights the functional role of the ASFV pE301R as a viral sliding clamp analog, but also facilitates the dissection of the complex replication mechanism of ASFV, which provides novel clues for developing antivirals against ASF.


Sujet(s)
Virus de la peste porcine africaine , Suidae , Animaux , Virus de la peste porcine africaine/génétique , Réplication virale , DNA-directed DNA polymerase , Cellules eucaryotes
2.
J Virol ; 97(10): e0082423, 2023 10 31.
Article de Anglais | MEDLINE | ID: mdl-37724880

RÉSUMÉ

IMPORTANCE: African swine fever (ASF) is an acute, hemorrhagic, and severe porcine infectious disease caused by African swine fever virus (ASFV). ASF outbreaks severely threaten the global pig industries and result in serious economic losses. No safe and efficacious commercial vaccine is currently available except in Vietnam. To date, large gaps in the knowledge concerning viral biological characteristics and immunoevasion strategies have hindered the ASF vaccine design. In this study, we demonstrate that pD129L negatively regulates the type I interferon (IFN) signaling pathway by interfering with the interaction of the transcriptional coactivator p300 and IRF3, thereby inhibiting the induction of type I IFNs. This study reveals a novel immunoevasion strategy employed by ASFV, shedding new light on the intricate mechanisms for ASFV to evade the host immune responses.


Sujet(s)
Virus de la peste porcine africaine , Peste porcine africaine , Protéine p300-E1A , Facteur-3 de régulation d'interféron , Interféron de type I , Animaux , Peste porcine africaine/virologie , Interféron de type I/métabolisme , Interféron bêta/métabolisme , Suidae , Facteurs de transcription/métabolisme , Vaccins/métabolisme , Protéine p300-E1A/métabolisme , Facteur-3 de régulation d'interféron/métabolisme , Échappement immunitaire
3.
J Virol ; 96(9): e0195721, 2022 05 11.
Article de Anglais | MEDLINE | ID: mdl-35412346

RÉSUMÉ

African swine fever is a lethal hemorrhagic disease of pigs caused by African swine fever virus (ASFV), which greatly threatens the pig industry in many countries. Deletion of virulence-associated genes to develop live attenuated ASF vaccines is considered to be a promising strategy. A recent study has revealed that the A137R gene deletion results in ASFV attenuation, but the underlying mechanism remains unknown. To elucidate the mechanism of the A137R gene regulating ASFV virulence, an ASFV mutant with the A137R gene deleted (ASFV-ΔA137R) was generated based on the wild-type ASFV HLJ/2018 strain (ASFV-WT). Using transcriptome sequencing analysis, we found that ASFV-ΔA137R induced higher type I interferon (IFN) production in primary porcine alveolar macrophages (PAMs) than did ASFV-WT. Overexpression of the A137R protein (pA137R) inhibited the activation of IFN-ß or IFN-stimulated response element. Mechanistically, pA137R interacts with TANK-binding kinase 1 (TBK1) and promotes the autophagy-mediated lysosomal degradation of TBK1, which blocks the nuclear translocation of interferon regulator factor 3, leading to decreased type I IFN production. Taken together, our findings clarify that pA137R negatively regulates the cGAS-STING-mediated IFN-ß signaling pathway via the autophagy-mediated lysosomal degradation of TBK1, which highlights the involvement of pA137R regulating ASFV virulence. IMPORTANCE African swine fever (ASF) is a lethal viral disease of pigs caused by African swine fever virus (ASFV). No commercial vaccines and antiviral treatments are available for the prevention and control of the disease. Several virulence-associated genes of ASFV have been identified, but the underlying attenuation mechanisms are not clear. Compared with the virulent parental ASFV, the A137R gene-deleted ASFV mutant promoted the expression of type I interferon (IFN) in primary porcine alveolar macrophages. Further analysis indicated that the A137R protein negatively regulated the cGAS-STING-mediated IFN-ß signaling pathway through targeting TANK-binding kinase 1 (TBK1) for autophagy-mediated lysosomal degradation. This study not only facilitates the understanding of ASFV immunoevasion strategies, but also provides new clues to the development of live attenuated ASF vaccines.


Sujet(s)
Virus de la peste porcine africaine , Autophagie , Interféron bêta , Protein-Serine-Threonine Kinases , Protéines virales , Peste porcine africaine , Virus de la peste porcine africaine/génétique , Animaux , Interféron bêta/métabolisme , Lysosomes/métabolisme , Macrophages alvéolaires/virologie , Protéines membranaires , Nucleotidyltransferases/métabolisme , Protein-Serine-Threonine Kinases/métabolisme , Suidae , Protéines virales/génétique , Virulence
4.
Front Microbiol ; 13: 839585, 2022.
Article de Anglais | MEDLINE | ID: mdl-35222345

RÉSUMÉ

Macrophages are professional antigen-presenting cells and serve as the first line of defense against invading pathogens. Macrophages are polarized toward the proinflammatory classical (M1) or anti-inflammatory alternative (M2) phenotype upon viral infections. M1-polarized macrophages exert critical roles in antiviral responses via different mechanisms. Within the long competitive history between viruses and hosts, viruses have evolved various immune evasion strategies, inhibiting macrophage acquisition of an antiviral phenotype, impairing the antiviral responses of activated macrophages, and/or exploiting macrophage phenotypes for efficient replication. This review focuses on the sophisticated regulation of macrophage polarization utilized by viruses and is expected to provide systematic insights into the regulatory mechanisms of macrophage polarization by viruses and further facilitate the design of therapeutic targets for antivirals.

5.
EBioMedicine ; 41: 156-166, 2019 Mar.
Article de Anglais | MEDLINE | ID: mdl-30833190

RÉSUMÉ

BACKGROUND: Activation of autocrine VEGF-VEGFR2 signalling in tumour cells activates cell proliferation, survival, and angiogenesis, all of which are crucial for tumour progression. Ovarian cancer-associated antigen 66 (OVA66) is now known to be overexpressed in multiple tumours and plays a role in tumour development, but the underlying mechanisms has not been fully investigated. METHODS: We employed ovarian and cervical cancer cells and mouse models to detect the role of OVA66 in angiogenesis, growth, and metastasis of cancer cells. Immunofluorescence and western blot were used to determine the function of OVA66 in regulating autocrine VEGF-VEGFR2 signalling. Immunohistochemistry and bioinformatics analysis were used to detect the correlation of OVA66 and VEGF expression. FINDINGS: OVA66 overexpression in the cancer cell lines promoted VEGF secretion, tumour growth and angiogenesis in vitro and in vivo. Conversely, shRNA-mediated OVA66 knockdown had the opposite effects. Mechanistically, OVA66 overexpression was found to boost an autocrine VEGF-VEGFR2 positive-feedback signalling loop in the tumour cells, leading to amplified effect of VEGF on tumour angiogenesis and proliferation and increased migration in vitro and in vivo, respectively. Finally, we identified a significant positive correlation between the expression levels of OVA66 and VEGF in ovarian and cervical cancer specimens, and found that OVA66 was significantly associated with advanced ovarian cancer. INTERPRETATION: We identify a novel function for OVA66 in regulating an autocrine VEGF-VEGFR2 feed-forward signalling loop that promotes tumour progression and angiogenesis. FUND: This work was supported by the National Natural Science Foundation of China (81602262); and Excellent Youth Scholar Program of Tongji University (2015KJ062).


Sujet(s)
Communication autocrine , Néovascularisation pathologique/métabolisme , Tumeurs de l'ovaire/métabolisme , Facteur de croissance endothéliale vasculaire de type A/métabolisme , Récepteur-2 au facteur croissance endothéliale vasculaire/métabolisme , Animaux , Antigènes néoplasiques/génétique , Antigènes néoplasiques/métabolisme , Femelle , Cellules HeLa , Humains , Souris , Néovascularisation pathologique/génétique , Néovascularisation pathologique/anatomopathologie , Tumeurs de l'ovaire/génétique , Tumeurs de l'ovaire/anatomopathologie , Transduction du signal
6.
Oncotarget ; 8(32): 52854-52865, 2017 Aug 08.
Article de Anglais | MEDLINE | ID: mdl-28881777

RÉSUMÉ

Ovarian cancer-associated antigen 12 (OVA12) was first identified in an ovarian carcinoma complementary DNA (cDNA) expression library and has been shown to play an important role in tumor growth. Here, we found that overexpression of OVA12 accelerated tumor growth in different tumor cells, whereas OVA12 depletion was associated with the opposite effect. Moreover, knocking down OVA12 led to a significant increase in the protein levels of p53, and the overexpression of OVA12 significantly decreased endogenous p53 levels. In addition, OVA12 stimulated p53 polyubiquitination and degradation by the proteasome and promoted tumor growth at least partially through the p53 pathway. Taken together, these results indicate that OVA12 is a negative regulator of p53 and that inhibition of OVA12 expression might serve as a therapeutic target to restore tumor suppression.

7.
Oncotarget ; 7(32): 51174-51183, 2016 Aug 09.
Article de Anglais | MEDLINE | ID: mdl-27203674

RÉSUMÉ

Originally identified as an E3 ligase regulating toll-like receptor (TLR) signaling, ring finger protein 216 (RNF216) also plays an essential role in autophagy, which is fundamental to cellular homeostasis. Autophagy dysfunction leads to an array of pathological events, including tumor formation. In this study, we found that RNF216 was upregulated in human colorectal cancer (CRC) tissues and cell lines, and was associated with progression of CRC. RNF216 promoted CRC cell proliferation and migration in vitro and in vivo, largely by enhancing proteasomal degradation of BECN1, a key autophagy regulator and tumor suppressor. RNF216 restricted CRC cell autophagy through BECN1 inhibition under nutritional starvation conditions. RNF216 knockdown increased the autophagy, limiting CRC cell proliferation and migration. Moreover, BECN1 knockdown or autophagy inhibition restored proliferation and migration of RNF216-knockdown CRC cells. Collectively, our results suggested that RNF216 promoted CRC cell proliferation and migration by negatively regulating BECN1-dependent autophagy. This makes RNF216 as a potential biomarker and novel therapeutic target for inhibiting CRC development and progression.


Sujet(s)
Autophagie/génétique , Bécline-1/physiologie , Mouvement cellulaire/génétique , Prolifération cellulaire/génétique , Tumeurs colorectales/génétique , Tumeurs colorectales/anatomopathologie , Ubiquitin-protein ligases/physiologie , Adulte , Sujet âgé , Sujet âgé de 80 ans ou plus , Animaux , Bécline-1/génétique , Marqueurs biologiques tumoraux/génétique , Cellules Caco-2 , Lignée cellulaire tumorale , Femelle , Régulation de l'expression des gènes tumoraux , Gènes suppresseurs de tumeur/physiologie , Cellules HT29 , Humains , Mâle , Souris , Souris de lignée BALB C , Souris nude , Adulte d'âge moyen , Ubiquitin-protein ligases/génétique
8.
Oncol Rep ; 35(6): 3293-302, 2016 Jun.
Article de Anglais | MEDLINE | ID: mdl-27109463

RÉSUMÉ

It has been widely suggested that mammosphere-forming cells from tumor cell lines or primary tumors represent the population of cancer stem cells (CSCs), which is supposed to lead to the failure of routine chemotherapy and the recurrence of the disease. However, it is still difficult to obtain CSCs from primary breast cancer for further investigation. We performed a modified culture system to generate mammosphere-forming cells derived from freshly isolated human breast cancer samples and the breast cancer cell line MCF-7. Cancer stem cell-like phenotypes such as CD44 and CD24 were measured by flow cytometry while alkaline phosphatase (AP) and mammaglobin (MGB1) expression was evaluated immunohistochemically. The expression levels of Klf4, Nanog, Oct4, Sox2 and mdr1 genes were analyzed by quantitative real­time PCR. Resistance to chemotherapeutic drugs was detected through the apoptosis assay upon drug treatments together with the detection of drug-resistant gene mdr1. The results revealed that we successfully obtained mammosphere­forming cells from the primary breast cancer in conditioned medium after 14 days of culture. Mammosphere-forming cells from primary breast cancer displayed a CD44hiCD24lo phenotype as well as positive AP and MGB1 reactivity. Stem cell-related genes such as Klf4, Nanog and Oct4 were detectably expressed in these cells. These cells formed tumor-like structures in the lymph nodes of nude mice, which were morphologically and histologically similar to breast cancer. Compared to the breast cancer cell line MCF-7 or mammosphere-forming cells from MCF-7 cells, the mammosphere-forming cells from the primary breast cancer exhibited resistance to three of four first-line chemotherapeutic drugs investigated through the induction of apoptosis, which was largely associated with the increased expression of drug-resistant gene mdr1 upon drug treatment. In conclusion, mammosphere-forming cells generated from the primary breast cancer exhibit CSC-like properties together with multiple drug resistance. Determination of the sensitivity of these primary cancer-derived mammosphere-forming cells to chemotherapeutic drugs may thus provide useful instructions for individualized therapy against the recurrence of breast cancer in the future.


Sujet(s)
Antinéoplasiques/pharmacologie , Tumeurs du sein/anatomopathologie , Antigènes CD24/métabolisme , Antigènes CD44/métabolisme , Cellules souches tumorales/métabolisme , Sous-famille B de transporteurs à cassette liant l'ATP/métabolisme , Animaux , Apoptose , Marqueurs biologiques tumoraux , Tumeurs du sein/traitement médicamenteux , Tumeurs du sein/métabolisme , Multirésistance aux médicaments , Résistance aux médicaments antinéoplasiques , Femelle , Humains , Facteur-4 de type Kruppel , Cellules MCF-7 , Souris nude , Cellules souches tumorales/effets des médicaments et des substances chimiques , Sphéroïdes de cellules , Tests d'activité antitumorale sur modèle de xénogreffe
9.
Nanoscale Res Lett ; 10: 103, 2015.
Article de Anglais | MEDLINE | ID: mdl-25852398

RÉSUMÉ

The two-dimensional (2D) triangle lattice air hole photonic crystal (PC) GaN-based light-emitting diodes (LED) with double-layer graphene transparent electrodes (DGTE) have been produced. The current spreading effect of the double-layer graphene (GR) on the surface of the PC structure of the LED has been researched. Specially, we found that the part of the graphene suspending over the air hole of the PC structure was of much higher conductivity, which reduced the average sheet resistance of the graphene transparent conducting electrode and improved the current spreading of the PC LED. Therefore, the work voltage of the DGTE-PC LED was obviously decreased, and the output power was greatly enhanced. The COMSOL software was used to simulate the current density distribution of the samples. The results show that the etching of PC structure results in the degradation of the current spreading and that the graphene transparent conducting electrode can offer an uniform current spreading in the DGTE-PC LED. PACS: 85.60.Jb; 68.65.Pq; 42.70.Qs.

10.
Cancer Lett ; 357(1): 141-151, 2015 Feb 01.
Article de Anglais | MEDLINE | ID: mdl-25444895

RÉSUMÉ

To achieve a better understanding of mechanisms that underlie carcinogenesis and to identify novel target molecules for diagnosis and therapy of carcinoma, we previously identified 24 distinct gene clones by immunoscreening of a cDNA library derived from an ovarian cancer patient through SEREX analysis. Among these genes we focused on a novel gene termed OVA12 and which putatively encodes a 114-amino-acid protein. In the present study, we found that OVA12 was ubiquitously overexpressed in diverse human tumor cell lines. Interestingly, we noticed that overexpression of OVA12 promoted proliferation of cancer cells in vitro and accelerated tumor growth in nude mice as compared to controls. Conversely, specific downregulation of OVA12 inhibited tumor cell proliferation and tumor growth both in vitro and in vivo. Furthermore, OVA12 inhibited 5-FU-induced apoptosis through specific upregulation of Mcl-1 and survivin. These results demonstrate that OVA12 is able to promote tumor growth, suggesting that this antigen might be a new potential target for development of cancer therapy.


Sujet(s)
Antigènes néoplasiques/métabolisme , Fluorouracil/antagonistes et inhibiteurs , Tumeurs/traitement médicamenteux , Tumeurs/métabolisme , Animaux , Antigènes néoplasiques/génétique , Apoptose/effets des médicaments et des substances chimiques , Lignée cellulaire tumorale , Prolifération cellulaire/effets des médicaments et des substances chimiques , Femelle , Fluorouracil/pharmacologie , Cellules HeLa , Humains , Souris , Souris de lignée BALB C , Souris nude , Tumeurs/anatomopathologie , Transfection , Tests d'activité antitumorale sur modèle de xénogreffe
11.
Shanghai Kou Qiang Yi Xue ; 23(3): 280-4, 2014 Jun.
Article de Chinois | MEDLINE | ID: mdl-25102867

RÉSUMÉ

PURPOSE: To investigate the differentiation and osteogenic activity of Naringin-induced bone marrow stromal cells (BMSCs) in dogs. METHODS: BMSCs were separated and cultured in vitro and identified by FCM. Then different concentration of Naringin (1×10⁻5, 1×10⁻6, 1×10⁻7, 1×10⁻8 and 1×10⁻9 mol/L) were added to cell culture media to induce BMSCs. The effect of Naringin on BMSCs was evaluated respectively by CCK-8 method and measuring the activity of alkaline phosphatase (ALP). The formation of nodules of calcium was detected by von Kossa staining. The data was analyzed with SPSS20.0 software package. RESULTS: The result of cell-surface marker displayed that the expression of CD34 and CD45 were negative while the expression of CD90 was positive. The values were 0.126%, 0.075% and 95.4%, respectively. Naringin could obviously promote cell proliferation, which exhibited the best effect on proliferation and osteogenic differentiation at concentration of 10-6 mol/L. Calcium nodule (von Kossa) staining was positive. CONCLUSIONS: Naringin-induced bone marrow stromal cells can be differentiated into osteoblasts. Naringin at the concentration of 10-6 mol/L can enhance the proliferation and osteogenic differentiation of BMSCs.


Sujet(s)
Flavanones/pharmacologie , Cellules souches mésenchymateuses , Ostéogenèse , Phosphatase alcaline , Animaux , Cellules de la moelle osseuse , Différenciation cellulaire , Prolifération cellulaire , Cellules cultivées , Chiens , Techniques in vitro , Ostéoblastes
12.
Carcinogenesis ; 35(7): 1573-81, 2014 Jul.
Article de Anglais | MEDLINE | ID: mdl-24667688

RÉSUMÉ

Ovarian cancer-associated antigen 66 (OVA66), also known as CML66 (GenBank Accession No. AF283301), was first identified in an ovarian carcinoma complementary DNA (cDNA) expression library and was shown to play a role in tumorigenesis. Here, we find that OVA66 influences tumorigenesis by regulating the type I insulin-like growth factor receptor (IGF-1R) signaling pathway. Stable knockdown of OVA66 in cancer cells attenuated phosphorylation of IGF-1R and extracellular signal-regulated kinase 1/2 (ERK1/2)-Hsp27; similarly, a higher level of p-IGF-1R and ERK1/2-Hsp27 signaling was also detected after OVA66 overexpression in HO8910 cells. In vivo knockdown of OVA66 both reduced tumor burden in nude mice and decreased phosphorylation of IGF-1R, ERK1/2 and hsp27. We blocked IGF-1R function both by small interfering RNA (siRNA) and with the chemical inhibitor Linsitinib (OSI-906). By either method, tumorigenesis was inhibited regardless of OVA66 expression; thus, mechanistically, IGF-1R, probably, lies downstream of OVA66 in cancer cells. We also found that OVA66 regulates expression of murine double minute 2 (MDM2); this attenuates ubiquitination of IGF-1R in response to IGF-1 stimulation and promotes active ERK1/2 signaling. Thus, we propose that combined overexpression of OVA66 and MDM2 promotes oncogenesis by enhancing activation of the IGF-1R-ERK1/2 signaling pathway.


Sujet(s)
Antigènes néoplasiques/métabolisme , Mouvement cellulaire , Prolifération cellulaire , Régulation de l'expression des gènes tumoraux , Mitogen-Activated Protein Kinases/métabolisme , Tumeurs/anatomopathologie , Tumeurs de l'ovaire/anatomopathologie , Récepteur IGF de type 1/métabolisme , Animaux , Antigènes néoplasiques/composition chimique , Antigènes néoplasiques/génétique , Apoptose , Technique de Western , Cycle cellulaire , Femelle , Humains , Techniques immunoenzymatiques , Souris , Souris de lignée BALB C , Souris nude , Invasion tumorale , Tumeurs/métabolisme , Tumeurs de l'ovaire/métabolisme , Analyse par réseau de protéines , Petit ARN interférent/génétique , Récepteur IGF de type 1/antagonistes et inhibiteurs , Récepteur IGF de type 1/génétique , Cellules cancéreuses en culture , Tests d'activité antitumorale sur modèle de xénogreffe
13.
PLoS One ; 9(3): e85705, 2014.
Article de Anglais | MEDLINE | ID: mdl-24633332

RÉSUMÉ

The tumor associated antigen OVA66 has been demonstrated to be highly expressed in malignant tumors and implicated in various cellular processes. To further elucidate its oncogenic character, we established an OVA66 stably overexpressed NIH3T3 cell line and a vector transfected control, named NIH3T3-flagOVA66 and NIH3T3-mock, respectively. NIH3T3-flagOVA66 cells showed faster cell cycling, proliferation, cell migration and more resistance to 5-fluorouracil-induced apoptosis. When NIH3T3-flagOVA66 and NIH3T3-mock cells were injected into nude mice for xenograft tumorigenicity assays, the NIH3T3-flagOVA66 cells formed tumors whereas no tumors were observed in mice inoculated with NIH3T3-mock cells. Analysis of PI3K/AKT and ERK1/2 MAPK signaling pathways by serum stimulation indicated hyperactivation of AKT and ERK1/2 in NIH3T3-flagOVA66 cells compared with NIH3T3-mock cells, while a decreased level of p-AKT and p-ERK1/2 were observed in OVA66 knocked down HeLa cells. To further validate if the p-AKT or p-ERK1/2 is essential for OVA66 induced oncogenic transformation, we treated the cells with the PI3K/AKT specific inhibitor LY294002 and the ERK1/2 MAPK specific inhibitor PD98059 and found either inhibitor can attenuate the cell colony forming ability in soft agar and the cell viability of NIH3T3-flagOVA66 cells, suggesting aberrantly activated AKT and ERK1/2 signaling be indispensible of the tumorigenic role of OVA66. Our results indicate that OVA66 is important in oncogenic transformation, promoting proliferation, cell migration and reducing apoptosis via hyperactivating PI3K/AKT and ERK1/2 MAPK signaling pathway. Thus, OVA66 might be a novel target for early detection, prevention and treatment of tumors in the future.


Sujet(s)
Antigènes néoplasiques/métabolisme , Transformation cellulaire néoplasique/métabolisme , Animaux , Antigènes néoplasiques/génétique , Transformation cellulaire néoplasique/effets des médicaments et des substances chimiques , 4H-1-Benzopyran-4-ones/pharmacologie , Flavonoïdes/pharmacologie , Cellules HeLa , Humains , Souris , Morpholines/pharmacologie , Cellules NIH 3T3 , Phosphatidylinositol 3-kinases/métabolisme , Inhibiteurs des phosphoinositide-3 kinases , Protéines proto-oncogènes c-akt/antagonistes et inhibiteurs , Protéines proto-oncogènes c-akt/métabolisme , Transduction du signal/effets des médicaments et des substances chimiques
14.
Int J Oncol ; 44(4): 1277-83, 2014 Apr.
Article de Anglais | MEDLINE | ID: mdl-24452693

RÉSUMÉ

Trastuzumab resistance is a challenging problem in ErbB2/HER2-positive breast cancers. Multiple mechanisms of resistance have been proposed and, thus, may require the development of more personalized therapies. In this study, we report the establishment of a mouse mammary cancer cell line, designated MT104T, obtained from spontaneous tumors in genetically engineered FVB/N-ErbB2/Neu-positive-PTEN-deficient mice. The critical molecular phenotype of MT104T cells was confirmed by genotyping and western blot analysis. This cell line was tumorigenic in immunologically intact syngeneic mice, forming tumors of generally similar histology as its origin. PTEN loss led to hyperactivation of Akt and conferred resistance to anti-ErbB2/Neu antibody treatment in MT104T cells. Addition of the Akt inhibitor triciribine (TCN) inhibited the viability of MT104T cells in a dose- and time-dependent manner as evaluated by MTT assay. ErbB2/Neu antibody and TCN combination treatment greatly induced apoptosis of MT104T cells as indicated by Annexin V-FITC staining. Moreover, this combination treatment also significantly reduced both Akt and Erk activities, which are responsible for the inhibitory effect on MT104T cells. Therefore, MT104T cells could represent an alternative model system to investigate the nature of ErbB2­positive breast cancer and for the experimental therapeutics studies of this disease. Our findings also suggest that combination of TCN may be a potential strategy for the treatment of trastu-zumab-resistant breast cancer mediated by PTEN loss or PI3K hyperactivation, which may facilitate the development of more personalized therapies for breast cancer patients.


Sujet(s)
Tumeurs mammaires de l'animal/traitement médicamenteux , Phosphohydrolase PTEN/génétique , Protéines proto-oncogènes c-akt/antagonistes et inhibiteurs , Récepteur ErbB-2/génétique , Ribonucléosides/pharmacologie , Animaux , Anticorps monoclonaux humanisés/pharmacologie , Antinéoplasiques/pharmacologie , Apoptose/effets des médicaments et des substances chimiques , Lignée cellulaire tumorale , Survie cellulaire/effets des médicaments et des substances chimiques , Résistance aux médicaments antinéoplasiques , Femelle , Tumeurs mammaires de l'animal/immunologie , Souris , Souris knockout , Phosphatidylinositol 3-kinases/biosynthèse , Récepteur ErbB-2/immunologie , Trastuzumab
15.
FEMS Yeast Res ; 13(4): 386-93, 2013 Jun.
Article de Anglais | MEDLINE | ID: mdl-23480635

RÉSUMÉ

The widespread use of azoles has led to increasing azole resistance among Candida albicans strains. One mechanism of azole resistance involves point mutations in the ERG11 gene, which encodes the target enzyme (cytochrome P450 lanosterol 14α-demethylase). In the present study, we amplified and sequenced the ERG11 gene of 23 C. albicans clinical isolates. Seventeen mutations encoding distinct amino acid substitutions were found, of which seven (K143Q, Y205E, A255V, E260V, N435V, G472R, and D502E) were novel. We further verified the contribution of the amino acid substitutions to azole resistance using site-directed mutagenesis of the ERG11 gene to recreate these mutations for heterologous expression in Saccharomyces cerevisiae. We observed that substitutions A114S, Y132H, Y132F, K143R, Y257H, and a new K143Q substitution contributed to significant increases (≧fourfold) in fluconazole and voriconazole resistance; changes in itraconazole resistance were not significant (≦twofold).


Sujet(s)
Antifongiques/pharmacologie , Azoles/pharmacologie , Candida albicans/effets des médicaments et des substances chimiques , Candida albicans/génétique , Cytochrome P-450 enzyme system/génétique , Résistance des champignons aux médicaments , Mutation faux-sens , Substitution d'acide aminé , Candida albicans/isolement et purification , Candidose/microbiologie , Cytochrome P-450 enzyme system/métabolisme , Analyse de mutations d'ADN , ADN fongique/composition chimique , ADN fongique/génétique , Humains , Mutagenèse dirigée , Protéines mutantes/génétique , Protéines mutantes/métabolisme , Analyse de séquence d'ADN
16.
Cancer Res ; 72(17): 4417-28, 2012 Sep 01.
Article de Anglais | MEDLINE | ID: mdl-22773664

RÉSUMÉ

Trastuzumab is an iconic rationally designed targeted therapy for HER2-positive breast cancers. However, the low response rate and development of resistance call for novel approaches for the treatment of patients. Here, we report that concurrent targeting of tumor cells and activation of T cells in the tumor microenvironment results in a synergistic inhibitory effect on tumor growth and overcomes resistance in two distinct PTEN loss-mediated trastuzumab-resistant mammary tumor mouse models. In vivo combination treatment with HER2/Neu antibody and Akt inhibitor triciribine effectively inhibited tumor growth in both models via inhibiting PI3K/AKT and mitogen-activated protein kinase signaling accompanied by increased T-cell infiltration in the tumor microenvironment. We showed that both CD8(+) and CD4(+) T cells were essential to the optimal antitumor effect of this combination treatment in an IFN-γ-dependent manner. Importantly, the antitumor activities of HER2/Neu antibody and triciribine combination treatment were further improved when coinhibitory receptor cytotoxic T-lymphocyte-associated antigen 4 was blocked to enhance the T-cell response. Our data indicate that multitargeted combinatorial therapies targeting tumor cells and concomitantly enhancing T-cell response in the tumor microenvironment could cooperate to exert maximal therapeutic activity, suggesting a promising clinical strategy for treating trastuzumab-resistant breast cancers and other advanced malignancies.


Sujet(s)
Anticorps monoclonaux humanisés/pharmacologie , Antinéoplasiques/pharmacologie , Tumeurs du sein/immunologie , Lymphocytes T/immunologie , Animaux , Tumeurs du sein/génétique , Tumeurs du sein/métabolisme , Antigène CTLA-4/antagonistes et inhibiteurs , Antigène CTLA-4/immunologie , Lignée cellulaire , Résistance aux médicaments antinéoplasiques/génétique , Synergie des médicaments , Activation enzymatique/génétique , Femelle , Expression des gènes , Humains , Interféron gamma/immunologie , Lymphocytes TIL/immunologie , Souris , Souris transgéniques , Phosphohydrolase PTEN/génétique , Phosphohydrolase PTEN/métabolisme , Inhibiteurs de protéines kinases/pharmacologie , Protéines proto-oncogènes c-akt/métabolisme , Récepteur ErbB-2/antagonistes et inhibiteurs , Récepteur ErbB-2/génétique , Ribonucléosides/pharmacologie , Trastuzumab , Microenvironnement tumoral/immunologie
17.
J Cell Mol Med ; 15(10): 2040-5, 2011 Oct.
Article de Anglais | MEDLINE | ID: mdl-21073654

RÉSUMÉ

Allogeneic umbilical cord blood haematopoietic stem cells (UCB-HSCs) can be transplanted into a host with the intact innate immunity with limited immuno-reaction, although the mechanisms remain unclear. The present studies aimed at investigating potential mechanisms of allogeneic UCB-HSCs escape from the cytolysis of natural killer (NK) cells. We compared UCB-HSCs ability to protect from NK-mediated cytotoxicity with peripheral blood or bone marrow haematopoietic stem cells (PB-HSCs and BM-HSCs). HSCs expressed lower levels of natural cytotoxicity receptor ligands including NKp30L, NKp44L and NKp46L than monocytes. Blocking these ligands respectively or in combination could increase the resistance of HSCs against NK cell mediated cytotoxicity. High expression of HLA-G was noticed on UCB-HSCs, rather than PB-HSCs or BM-HSCs, whereas blockade of HLA-G significantly elevated NK cell mediated cytolysis to UCB-HSCs. Thus, we conclude that natural cytotoxicity receptors and HLA-G on HSCs may contribute to the escape from NK cells, and activate and inhibitory NK cell receptors and their ligands can be novel therapeutic targets in cell transplantation.


Sujet(s)
Sang foetal/cytologie , Antigènes HLA-G/métabolisme , Cellules souches hématopoïétiques/cytologie , Cellules tueuses naturelles/métabolisme , Récepteurs de déclenchement de cytotoxicité naturelle/métabolisme , Cytotoxicité immunologique , Sang foetal/métabolisme , Antigènes HLA-G/génétique , Transplantation de cellules souches hématopoïétiques , Cellules souches hématopoïétiques/métabolisme , Humains , Immunité innée , Cellules tueuses naturelles/cytologie , Monocytes/cytologie , Monocytes/métabolisme
18.
BMC Cancer ; 10: 184, 2010 May 07.
Article de Anglais | MEDLINE | ID: mdl-20459642

RÉSUMÉ

BACKGROUND: Insulin-like growth factor-I receptor (IGFIR) has been shown to regulate the tumor development. The objective of the current study is to determine the association of IGFIR with lymph node metastasis and to explore the related mechanism in human colorectal cancer in clinic. METHODS: In a random series of 98 colorectal cancer patients, the expressions of IGFIR, vascular endothelial growth factor (VEGF) and VEGF-C were investigated by immunohistochemistry, and the association of these expressions with lymph node metastasis was statistically analyzed. The expressions of VEGF and VEGF-C in colorectal cancer cells stimulated with IGF-I were also examined by real-time quantitative reverse transcription-polymerase chain reaction and enzyme-linked immunosorbent assay. RESULTS: Higher rates of IGFIR (46%), VEGF (53%), and VEGF-C (46%) expression were found in colorectal cancer tissues than in normal and colorectal adenoma tissues. These expressions were significantly associated with clinicopathologic factors and lymph node status. We also found the concomitant high expressions of IGFIR/VEGF (P < 0.001) and IGFIR/VEGF-C (P = 0.001) had a stronger correlation with lymph node metastasis than did each alone or both low expressions. In addition, IGF-I could effectively induce the VEGF and VEGF-C mRNA expression and protein secretion in colorectal cancer cells expressing IGFIR molecules. Moreover, Patients who had strong staining for IGFIR, VEGF and VEGF-C showed significantly less favorable survival rates compared with patients who had low staining for these molecules (P < 0.001). The survival rates of patients who were both high expression of IGFIR/VEGF and IGFIR/VEGF-C also were significantly lower compared with patients who were negative or one of high expression of these molecules (P < 0.001). CONCLUSIONS: Together the findings indicated for the first time that simultaneous examination of the expressions of IGFIR, VEGF and VEGF-C will benefit the diagnosis of lymph node metastasis in order to assay the prognosis and determine the treatment strategy in patients with colorectal cancer undergoing surgery.


Sujet(s)
Tumeurs colorectales/métabolisme , Tumeurs colorectales/anatomopathologie , Récepteur IGF de type 1/biosynthèse , Facteur de croissance endothéliale vasculaire de type A/biosynthèse , Facteur de croissance endothéliale vasculaire de type C/biosynthèse , Humains , Immunohistochimie , Facteur de croissance IGF-I/métabolisme , Facteur de croissance IGF-I/pharmacologie , Métastase lymphatique
19.
Cancer Lett ; 295(1): 17-26, 2010 Sep 01.
Article de Anglais | MEDLINE | ID: mdl-20202744

RÉSUMÉ

We previously found that dendritic cell (DC) precursors could be recruited into the peripheral blood of B6 mice by administration of macrophage inflammatory protein (MIP)-1alpha. These MIP-1alpha-recruited DCs could induce anti-tumor protective immunity when pulsed with tumor cell lysate. In this study, MIP-1alpha-recruited DCs could not effectively suppress preestablished tumor when pulsed with B16 tumor cell lysate. However, inoculation with these DCs expressing MAGE-1 induced an anti-tumor immunity against preestablished solid and metastatic tumor from B16-MAGE-1 cells. These MIP-1alpha-recruited DCs expressed higher level of CCR7 and displayed a more significant chemotactic response toward secondary lymphoid tissue. Therefore, they are superior in the induction of cytotoxic T lymphocytes and the inhibition of tumor development and metastasis than bone marrow-derived DCs. This study established a novel approach to the treatment of preestablished solid and metastatic tumors using MIP-1alpha-recruited DCs transduced with tumor antigen gene.


Sujet(s)
Vaccins anticancéreux/usage thérapeutique , Chimiokine CCL3/pharmacologie , Cellules dendritiques/immunologie , Immunothérapie , Mélanome expérimental/immunologie , Mélanome expérimental/thérapie , Animaux , Antigènes néoplasiques/génétique , Vaccins anticancéreux/immunologie , Lignée cellulaire tumorale , Chimiotaxie , Cytotoxicité immunologique , Femelle , Tumeurs du poumon/secondaire , Tumeurs du poumon/thérapie , Activation des lymphocytes , Mélanome expérimental/prévention et contrôle , Mélanome expérimental/secondaire , Antigènes spécifiques du mélanome , Souris , Souris de lignée C57BL , Protéines tumorales/génétique , Récepteurs CCR7/métabolisme , Protéines recombinantes/pharmacologie , Lymphocytes T cytotoxiques/immunologie , Transduction génétique
20.
Cancer Immunol Immunother ; 59(2): 279-91, 2010 Feb.
Article de Anglais | MEDLINE | ID: mdl-19727719

RÉSUMÉ

Till now, little is known about the effects of chemotherapy on the immunity of cancer patients and the ideal timing ("window" period) for immunotherapy combined with chemotherapy. In this study, we addressed the immunogenicity of apoptotic ovarian cancer cells induced by paclitaxel and carboplatin, the immunologic aspects in ovarian cancer patients under chemotherapy, and the CTL response when CD8(+) T cells were stimulated with tumor antigen in the "window" period. The immunogenicity of apoptotic ovarian cancer cells was detected first. Then, blood samples from each ovarian cancer patient were obtained before (S(0)) and at days 5-7 (S(1)), days 12-14 (S(2)) and days 25-28 (S(3)) after chemotherapy. The proportions of immunocyte subsets and the function of NK cells were studied. We found that apoptotic ovarian cancer cells elicited a powerful CTL response with antitumor activity in vitro. The proportions of CD3(+) T cells, CD4(+) T cells and the ratio of CD4(+) to CD8(+) cells did not change significantly on S(1), S(2) and S(3), compared to S(0), whereas the percentage of Treg cells decreased remarkably on S(2). The proportions of Th1, Tc1, CD45RO memory T, NKT cells and the ratio of Tc1 to Tc2 cells increased significantly on S(2). IFN-gamma secreting CD8(+) T cells also increased remarkably on S(2), especially when CD8(+) T cells were stimulated with autologous tumor antigen. From our point of view, chemotherapy induces temporary immune reconstitution and augments anti-tumor immune response. It is probable that the "window" period of days 12-14 after chemotherapy provides the best opportunity for immunotherapy.


Sujet(s)
Antinéoplasiques/usage thérapeutique , Carboplatine/usage thérapeutique , Tumeurs de l'ovaire/traitement médicamenteux , Tumeurs de l'ovaire/immunologie , Paclitaxel/usage thérapeutique , Lymphocytes T cytotoxiques/immunologie , Adulte , Apoptose/immunologie , Femelle , Humains , Immunothérapie , Numération des lymphocytes , Adulte d'âge moyen , Tumeurs de l'ovaire/anatomopathologie
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE