Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 113
Filtrar
1.
Mol Cell Biochem ; 477(4): 1041-1052, 2022 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-34994923

RESUMO

Cytotoxic chemotherapy dominates the field of cancer treatment. Consequently, anticancer phytochemicals are largely screened on the basis of their cytotoxicity towards cancer cells which are achieved at higher doses, leading to various toxic side effects. Some phytochemicals also showed pro-carcinogenic effects at certain doses. The concept of hormesis has taught us to look into biphasic responses of phytochemicals in a more systematic way. Interestingly, the monoterpenoid alcohol, linalool, also has been reported to display both anti-oxidant and pro-oxidant properties, which prompted us to explore a probable biphasic effect on cancer cells. Cytotoxicity of various concentrations of linalool (0.1-4 mM) was tested on B16F10 murine melanoma cell line, and two sub-lethal concentrations (0.4 and 0.8 mM) were selected for further experiments. 0.4 mM linalool inhibited angiogenesis and metastasis, while 0.8 mM increased them. Similarly, B16F10 cell migration, invasion, and epithelial-mesenchymal transition markers also showed inhibition and induction with lower and higher linalool concentrations, respectively. Chorioallantoic membrane assay, scratch wound assay, and Boyden's chamber were used to analyze angiogenesis and metastasis. Expression of molecular markers such as vascular endothelial growth factor (VEGF) and its receptor phosphorylated VEGF receptor II (p-VEGFRII or p-Flk-1), Hypoxia-inducible factor-1 α (HIF-1α), E-cadherin, and vimentin were detected using Western blot, ELISA, PCR, qPCR, and immunofluorescence. Finally, ChIP assay was performed to evaluate HIF-1α association with VEGF promoter. Interestingly, measurement of intracellular reactive oxygen species at the selected concentrations of linalool using DCFDA in a flow cytometer showed that the phytochemical induced significant amount of ROS at 0.8 mM. This work sheds light on bimodal dose-response relationship exhibited by dietary phytochemicals like linalool, and it should be taken into consideration to elicit a desirable therapeutic effect.


Assuntos
Monoterpenos Acíclicos/farmacologia , Melanoma Experimental , Neovascularização Patológica , Animais , Embrião de Galinha , Células HEK293 , Humanos , Melanoma Experimental/sangue , Melanoma Experimental/tratamento farmacológico , Melanoma Experimental/metabolismo , Melanoma Experimental/patologia , Camundongos , Metástase Neoplásica , Neovascularização Patológica/tratamento farmacológico , Neovascularização Patológica/metabolismo , Neovascularização Patológica/patologia
2.
Immunotherapy ; 14(2): 115-133, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-34783257

RESUMO

Aim: The aim of this work is to utilize a gene expression procedure to safely express systemic IL-12 and evaluate its effects in mouse tumor models. Materials & methods: Secondary lymphoid organs and tumors from EL4 and B16 tumor-bearing mice were analyzed by supervised and unsupervised methods. Results: IL-12 cDNA induced systemic IL-12 protein levels lower than the tolerated dose in patients. Control of tumor growth was observed in subcutaneous B16 and EL4 tumors. Systemic IL-12 expression induced a higher frequency of both total tumor-infiltrated CD45+ cells and proliferative IFN-γ+CD8+ T cells along with a lower frequency of CD4+FOXP3+ and CD11b+Gr-1+ cells. Conclusion: This approach characterizes the systemic effects of IL-12, helping to improve treatment of metastases or solid tumors.


Lay abstract IL-12 has emerged as a potent cytokine in mediating antitumor activity in preclinical models of cancer. However, this antitumor response has not yet been translated into the clinic because of toxic side effects. The aim of our work is to analyze the effects of IL-12 in mouse tumor models. We demonstrate that one injection of IL-12 cDNA can induce systemic IL-12 levels in serum even lower than the tolerated dose in patients. At this dose, an efficient control of tumor growth can be observed. We found a higher frequency of both total tumor-infiltrated leukocytes and IFN-γ-producing CD8+ T cells along with a lower frequency of regulatory CD4+FOXP3+ and CD11b+Gr1+ cells. Our work demonstrates that IL-12 cDNA can safely be used to treat cancer.


Assuntos
Adjuvantes Imunológicos/uso terapêutico , DNA Complementar/sangue , Interleucina-12/uso terapêutico , Linfoma/tratamento farmacológico , Melanoma Experimental/tratamento farmacológico , Animais , Modelos Animais de Doenças , Expressão Gênica , Interleucina-12/sangue , Linfoma/sangue , Linfoma/imunologia , Melanoma Experimental/sangue , Melanoma Experimental/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Transdução de Sinais
3.
J Immunol ; 206(10): 2412-2419, 2021 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-33911005

RESUMO

Sepsis induces significant immune dysregulation characterized by lymphocyte apoptosis and alterations in the cytokine milieu. Because cancer patients exhibit a 10-fold greater risk of developing sepsis compared with the general population, we aimed to understand how pre-existing malignancy alters sepsis-induced immune dysregulation. To address this question, we assessed the impact of tumor-specific CD8+ T cells on the immune response in a mouse model of cecal ligation and puncture (CLP)-induced sepsis. Tumor-bearing animals containing Thy1.1+ tumor-specific CD8+ T cells were subjected to CLP, and groups of animals received anti-Thy1.1 mAb to deplete tumor-specific CD8+ T cells or isotype control. Results indicated that depleting tumor-specific T cells significantly improved mortality from sepsis. The presence of tumor-specific CD8+ T cells resulted in increased expression of the 2B4 coinhibitory receptor and increased apoptosis of endogenous CD8+ T cells. Moreover, tumor-specific T cells were not reduced in number in the tumors during sepsis but did exhibit impaired IFN-γ production in the tumor, tumor draining lymph node, and spleen 24 h after CLP. Our research provides novel insight into the mechanisms by which pre-existing malignancy contributes to increased mortality during sepsis.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Imunidade , Neoplasias Pulmonares/complicações , Neoplasias Pulmonares/imunologia , Melanoma Experimental/complicações , Melanoma Experimental/imunologia , Sepse/complicações , Sepse/imunologia , Neoplasias Cutâneas/complicações , Neoplasias Cutâneas/imunologia , Animais , Apoptose/imunologia , Linhagem Celular Tumoral , Citocinas/sangue , Interferon gama/metabolismo , Neoplasias Pulmonares/sangue , Linfonodos/imunologia , Masculino , Melanoma Experimental/sangue , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Fenótipo , Sepse/sangue , Sepse/mortalidade , Neoplasias Cutâneas/sangue , Baço/imunologia , Antígenos Thy-1/genética
4.
Sci Rep ; 11(1): 5798, 2021 03 11.
Artigo em Inglês | MEDLINE | ID: mdl-33707612

RESUMO

Apelin, a ligand of the APJ receptor, is overexpressed in several human cancers and plays an important role in tumor angiogenesis and growth in various experimental systems. We investigated the role of apelin signaling in the malignant behavior of cutaneous melanoma. Murine B16 and human A375 melanoma cell lines were stably transfected with apelin encoding or control vectors. Apelin overexpression significantly increased melanoma cell migration and invasion in vitro, but it had no impact on its proliferation. In our in vivo experiments, apelin significantly increased the number and size of lung metastases of murine melanoma cells. Melanoma cell proliferation rates and lymph and blood microvessel densities were significantly higher in the apelin-overexpressing pulmonary metastases. APJ inhibition by the competitive APJ antagonist MM54 significantly attenuated the in vivo pro-tumorigenic effects of apelin. Additionally, we detected significantly elevated circulating apelin and VEGF levels in patients with melanoma compared to healthy controls. Our results show that apelin promotes blood and lymphatic vascularization and the growth of pulmonary metastases of skin melanoma. Further studies are warranted to validate apelin signaling as a new potential therapeutic target in this malignancy.


Assuntos
Apelina/efeitos adversos , Neoplasias Pulmonares/irrigação sanguínea , Neoplasias Pulmonares/secundário , Linfangiogênese , Melanoma Experimental/patologia , Neovascularização Patológica/patologia , Animais , Estudos de Casos e Controles , Linhagem Celular Tumoral , Movimento Celular , Proliferação de Células , Feminino , Humanos , Neoplasias Pulmonares/sangue , Masculino , Melanoma Experimental/sangue , Camundongos , Pessoa de Meia-Idade , Invasividade Neoplásica , Neovascularização Patológica/sangue , Fator A de Crescimento do Endotélio Vascular/sangue
5.
Bull Exp Biol Med ; 169(2): 254-257, 2020 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-32651814

RESUMO

We studied the effects of cathepsins produced by immunocompetent cells of intact mice, mice with B16 melanoma, mice with removed B16 melanoma, and mice with removed Ehrlich carcinoma on the growth of B16 melanoma. Incubation of B16 melanoma cells with cathepsins from immunocompetent cells of intact mice, mice with B16 melanoma, and mice with removed Ehrlich carcinoma did not affect tumor growth. Incubation of B16 melanoma cells with cathepsin from immunocompetent cells of mice with removed B16 melanoma was followed by complete loss of malignancy by these cells. Melanoma cells formed no tumor node within at least 1 year after transplantation, though exhibited high proliferative activity and formed pigmented nevi. The formation of a nevus by B16 melanoma cells is described for the first time. The existence of a mechanism regulating proliferation of malignant cells in the tumor-bearing host was hypothesized. Studies of this mechanism are expected to promote the development of new methods for the treatment of tumors.


Assuntos
Catepsinas/metabolismo , Melanócitos/patologia , Melanoma Experimental/metabolismo , Melanoma Experimental/patologia , Animais , Proliferação de Células/efeitos dos fármacos , Proliferação de Células/genética , Transformação Celular Neoplásica/efeitos dos fármacos , Transformação Celular Neoplásica/genética , Feminino , Melanoma Experimental/sangue , Camundongos , Camundongos Endogâmicos C57BL , Serpinas/sangue
6.
J Immunother Cancer ; 8(1)2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-32217757

RESUMO

BACKGROUND: CD6 is a lymphocyte surface co-receptor physically associated with the T-cell receptor (TCR)/CD3 complex at the center of the immunological synapse. There, CD6 assists in cell-to-cell contact stabilization and modulation of activation/differentiation events through interaction with CD166/ALCAM (activated leukocyte cell adhesion molecule), its main reported ligand. While accumulating evidence is attracting new interest on targeting CD6 for therapeutic purposes in autoimmune disorders, little is known on its potential in cancer. In an attempt to elucidate the in vivo relevance of blocking CD6-mediated interactions in health and disease, we explored the consequences of expressing high circulating levels of a soluble form CD6 (sCD6) as a decoy receptor. METHODS: High sCD6 serum levels were achieved by using transgenic C57BL/6 mice expressing human sCD6 under the control of lymphoid-specific transcriptional elements (shCD6LckEµTg) or wild type either transduced with hepatotropic adeno-associated virus coding for mouse sCD6 or undergoing repeated infusions of recombinant human sCD6 protein. Characterization of sCD6-induced changes was performed by ex vivo flow cytometry and functional analyses of mouse lymphoid organ cells. The in vivo relevance of those changes was explored by challenging mice with subcutaneous or metastatic tumors induced by syngeneic cancer cells of different lineage origins. RESULTS: Through a combination of in vitro and in vivo studies, we show that circulating sCD6 expression induces defective regulatory T cell (Treg) generation and function, decreased CD166/ALCAM-mediated tumor cell proliferation/migration and impaired galectin-induced T-cell apoptosis, supporting the fact that sCD6 modulates antitumor lymphocyte effector function and tumorigenesis. Accordingly, sCD6 expression in vivo resulted in delayed subcutaneous tumor growth and/or reduced metastasis on challenge of mice with syngeneic cancer cells. CONCLUSIONS: Evidence is provided for the disruption of CD6 receptor-ligand interactions as a feasible immunomodulatory approach in cancer.


Assuntos
Antígenos CD/sangue , Antígenos de Diferenciação de Linfócitos T/sangue , Neoplasias Pulmonares/imunologia , Linfoma de Células T/imunologia , Melanoma Experimental/imunologia , Sarcoma Experimental/imunologia , Linfócitos T Reguladores/imunologia , Molécula de Adesão de Leucócito Ativado/imunologia , Molécula de Adesão de Leucócito Ativado/metabolismo , Animais , Antígenos CD/administração & dosagem , Antígenos CD/biossíntese , Antígenos CD/genética , Antígenos de Diferenciação de Linfócitos T/administração & dosagem , Antígenos de Diferenciação de Linfócitos T/biossíntese , Antígenos de Diferenciação de Linfócitos T/genética , Apoptose/fisiologia , Diferenciação Celular/fisiologia , Linhagem Celular Tumoral , Feminino , Humanos , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/secundário , Linfoma de Células T/metabolismo , Masculino , Melanoma Experimental/sangue , Melanoma Experimental/patologia , Melanoma Experimental/terapia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Proteínas Recombinantes/administração & dosagem , Proteínas Recombinantes/sangue , Proteínas Recombinantes/genética , Sarcoma Experimental/sangue , Sarcoma Experimental/patologia , Linfócitos T Reguladores/metabolismo
7.
Nano Lett ; 20(1): 372-383, 2020 01 08.
Artigo em Inglês | MEDLINE | ID: mdl-31840517

RESUMO

Angiogenesis is the hallmark of melanoma that nurtures the tumor microenvironment (TME) for rapid tumor progression. Vessel normalization could benefit melanoma treatment through TME reconstruction, while its limited duration and extent are still the drag. Herein, two kinds of look-like nanodrugs, called Gemini-like nanodrugs (GLnano), were constructed separately with the same scaffold of antiangiogenic low molecular weight heparin (LMWH) and mixed upon administration in vivo. For one, doxorubicin (DOX) was encapsulated into LMWH-chrysin nanodrug (LCY) with DSPE-PEG-anisamide decoration (D-LCA nanodrugs) for active targeting and direct cell killing toward melanoma cells. For another, matrix metalloproteinases (MMPs)-sensitive peptide was conjugated to LMWH to encapsulate celecoxib (Cel) (C-Lpep nanodrugs), disassembling in TME by MMPs and releasing Cel for M2-to-M1 reprogramming of tumor-associated macrophages. Our results showed that GLnano could remarkably elongate the vessel normalization window up to 12 days with the highest pericyte coverage of nearly 75%, compared to only 4 days by LCY monotherapy. Furthermore, GLnano could spontaneously form the "treatment-delivery" loop to promote nanodrugs toward deep tumor regions, leading to a potent tumor inhibition, metastasis prevention, and overall TME improvements.


Assuntos
Doxorrubicina , Sistemas de Liberação de Medicamentos , Heparina de Baixo Peso Molecular , Melanoma Experimental , Nanopartículas , Neovascularização Patológica , Microambiente Tumoral/efeitos dos fármacos , Animais , Doxorrubicina/química , Doxorrubicina/farmacocinética , Doxorrubicina/farmacologia , Heparina de Baixo Peso Molecular/química , Heparina de Baixo Peso Molecular/farmacocinética , Heparina de Baixo Peso Molecular/farmacologia , Melanoma Experimental/sangue , Melanoma Experimental/tratamento farmacológico , Melanoma Experimental/metabolismo , Melanoma Experimental/patologia , Camundongos , Nanopartículas/química , Nanopartículas/uso terapêutico , Neovascularização Patológica/tratamento farmacológico , Neovascularização Patológica/metabolismo , Neovascularização Patológica/patologia , Células RAW 264.7
8.
J Immunother Cancer ; 7(1): 114, 2019 04 26.
Artigo em Inglês | MEDLINE | ID: mdl-31027511

RESUMO

BACKGROUND: Induction of strong T cell responses, in particular cytotoxic T cells, is a key for the generation of efficacious therapeutic cancer vaccines which yet, remains a major challenge for the vaccine developing world. Here we demonstrate that it is possible to harness the physiological properties of the lymphatic system to optimize the induction of a protective T cell response. Indeed, the lymphatic system sharply distinguishes between nanoscale and microscale particles. The former reaches the fenestrated lymphatic system via diffusion, while the latter either need to be transported by dendritic cells or form a local depot. METHODS: Our previously developed cucumber-mosaic virus-derived nanoparticles termed (CuMVTT-VLPs) incorporating a universal Tetanus toxoid epitope TT830-843 were assessed for their draining kinetics using stereomicroscopic imaging. A nano-vaccine has been generated by coupling p33 epitope as a model antigen to CuMVTT-VLPs using bio-orthogonal Cu-free click chemistry. The CuMVTT-p33 nano-sized vaccine has been next formulated with the micron-sized microcrystalline tyrosine (MCT) adjuvant and the formed depot effect was studied using confocal microscopy and trafficking experiments. The immunogenicity of the nanoparticles combined with the micron-sized adjuvant was next assessed in an aggressive transplanted murine melanoma model. The obtained results were compared to other commonly used adjuvants such as B type CpGs and Alum. RESULTS: Our results showed that CuMVTT-VLPs can efficiently and rapidly drain into the lymphatic system due to their nano-size of ~ 30 nm. However, formulating the nanoparticles with the micron-sized MCT adjuvant of ~ 5 µM resulted in a local depot for the nanoparticles and a longer exposure time for the immune system. The preclinical nano-vaccine CuMVTT-p33 formulated with the micron-sized MCT adjuvant has enhanced the specific T cell response in the stringent B16F10p33 murine melanoma model. Furthermore, the micron-sized MCT adjuvant was as potent as B type CpGs and clearly superior to the commonly used Alum adjuvant when total CD8+, specific p33 T cell response or tumour protection were assessed. CONCLUSION: The combination of nano- and micro-particles may optimally harness the physiological properties of the lymphatic system. Since the nanoparticles are well defined virus-like particles and the micron-sized adjuvant MCT has been used for decades in allergen-specific desensitization, this approach may readily be translated to the clinic.


Assuntos
Adjuvantes Imunológicos/administração & dosagem , Vacinas Anticâncer/imunologia , Melanoma Experimental/terapia , Nanopartículas/administração & dosagem , Vacinas de Partículas Semelhantes a Vírus/imunologia , Animais , Vacinas Anticâncer/administração & dosagem , Cucumovirus/imunologia , Feminino , Imunogenicidade da Vacina , Melanoma Experimental/sangue , Melanoma Experimental/imunologia , Camundongos , Tamanho da Partícula , Fragmentos de Peptídeos/imunologia , Linfócitos T/imunologia , Toxoide Tetânico/imunologia , Tirosina/administração & dosagem , Vacinas de Partículas Semelhantes a Vírus/administração & dosagem
9.
Cell Metab ; 30(1): 143-156.e5, 2019 07 02.
Artigo em Inglês | MEDLINE | ID: mdl-31031094

RESUMO

Tumor-infiltrating T cells often lose their effector function; however, the mechanisms are incompletely understood. We report that cholesterol in the tumor microenvironment induces CD8+ T cell expression of immune checkpoints and exhaustion. Tumor tissues enriched with cholesterol and cholesterol content in tumor-infiltrating CD8+ T cells were positively and progressively associated with upregulated T cell expression of PD-1, 2B4, TIM-3, and LAG-3. Adoptively transferred CD8+ T cells acquired cholesterol, expressed high levels of immune checkpoints, and became exhausted upon entering a tumor. Tumor culture supernatant or cholesterol induced immune checkpoint expression by increasing endoplasmic reticulum (ER) stress in CD8+ T cells. Consequently, the ER stress sensor XBP1 was activated and regulated PD-1 and 2B4 transcription. Inhibiting XBP1 or reducing cholesterol in CD8+ T cells effectively restored antitumor activity. This study reveals a mechanism underlying T cell exhaustion and suggests a new strategy for restoring T cell function by reducing cholesterol to enhance T cell-based immunotherapy.


Assuntos
Linfócitos T CD8-Positivos/metabolismo , Colesterol/sangue , Microambiente Tumoral/fisiologia , Animais , Western Blotting , Citometria de Fluxo , Humanos , Imunoprecipitação , Imunoterapia , Melanoma Experimental/sangue , Camundongos , Receptor de Morte Celular Programada 1/metabolismo , Reação em Cadeia da Polimerase em Tempo Real
10.
Biomaterials ; 188: 118-129, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30343255

RESUMO

The prominent discrepancy between the significant investment towards plasma biomarker discovery and the very low number of biomarkers currently in clinical use stresses the need for discovery technologies. The discovery of protein biomarkers present in human blood by proteomics is tremendously challenging, owing to the large dynamic concentration range of blood proteins. Here, we describe the use of blood-circulating lipid-based nanoparticles (NPs) as a scavenging tool to comprehensively analyse the blood proteome. We aimed to exploit the spontaneous interaction of NPs with plasma proteins once injected in the bloodstream, known as 'protein corona', in order to facilitate the capture of tumor-specific molecules. We employed two different tumor models, a subcutaneous melanoma model (B16-F10) and human lung carcinoma xenograft model (A549) and comprehensively compared by mass spectrometry the in vivo protein coronas formed onto clinically used liposomes, intravenously administered in healthy and tumor-bearing mice. The results obtained demonstrated that blood-circulating liposomes surface-capture and amplify a wide range of different proteins including low molecular weight (MW) and low abundant tumor specific proteins (intracellular products of tissue leakage) that could not be detected by plasma analysis, performed in comparison. Most strikingly, the NP (liposomal) corona formed in the xenograft model was found to consist of murine host response proteins, as well as human proteins released from the inoculated and growing human cancer cells. This study offers direct evidence that the in vivo NP protein corona could be deemed as a valuable tool to enrich the blood proteomic analysis and to allow the discovery of potential biomarkers in experimental disease models.


Assuntos
Biomarcadores Tumorais/sangue , Proteínas Sanguíneas/análise , Lipossomos/metabolismo , Neoplasias Pulmonares/sangue , Melanoma Experimental/sangue , Coroa de Proteína/análise , Células A549 , Animais , Biomarcadores Tumorais/metabolismo , Proteínas Sanguíneas/metabolismo , Feminino , Humanos , Lipossomos/sangue , Neoplasias Pulmonares/metabolismo , Melanoma Experimental/metabolismo , Camundongos Endogâmicos C57BL , Nanopartículas/metabolismo , Coroa de Proteína/metabolismo
11.
PLoS One ; 13(11): e0206827, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30383838

RESUMO

Hyperuricaemia is associated with various metabolic dysfunctions including obesity, type 2 diabetes mellitus, hypertension and in general metabolic syndrome, which are all associated with increased risk of cancer. However, the direct association between elevated uricemia and cancer mortality still remains unclear. In this study, we used a mouse model of hyperuricemia, the Urahplt2/plt2 (PLT2) mouse, to investigate the effect of high uric acid levels on anti-tumor immune responses and tumor growth. In normo-uricaemic C57BL/6 mice injected with B16 melanomas, immunotherapy by treatment with Poly I:C at the tumor site delayed tumor growth compared to PBS treatment. In contrast, Poly I:C-treated hyper-uricaemic PLT2 mice were unable to delay tumor growth. Conventional and monocyte-derived dendritic cells in the tumor-draining lymph nodes (dLN) of C57BL/6 and PLT2 mice were similarly increased after Poly I:C immunotherapy, and expressed high levels of CD40 and CD86. CD8+ T cells in the tumor-dLN and tumor of both WT and PLT2 mice were also increased after Poly I:C immunotherapy, and were able to secrete increased IFNγ upon in vitro restimulation. Surprisingly, tumor-specific CD8+ T cells in dLN were less abundant in PLT2 mice compared to C57BL/6, but showed a greater ability to proliferate even in the absence of cognate antigen. These data suggest that hyperuricaemia may affect the functionality of CD8+ T cells in vivo, leading to dysregulated T cell proliferation and impaired anti-tumor activity.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Hiperuricemia/imunologia , Indutores de Interferon/farmacologia , Melanoma Experimental/imunologia , Poli I-C/farmacologia , Neoplasias Cutâneas/imunologia , Amidoidrolases/genética , Amidoidrolases/metabolismo , Animais , Carcinogênese/imunologia , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Células Dendríticas/imunologia , Feminino , Humanos , Hiperuricemia/sangue , Hiperuricemia/genética , Hiperuricemia/metabolismo , Imunidade Celular/efeitos dos fármacos , Imunidade Celular/imunologia , Imunoterapia/métodos , Indutores de Interferon/uso terapêutico , Linfonodos/citologia , Linfonodos/imunologia , Masculino , Melanoma Experimental/sangue , Melanoma Experimental/tratamento farmacológico , Melanoma Experimental/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Poli I-C/uso terapêutico , Neoplasias Cutâneas/sangue , Neoplasias Cutâneas/tratamento farmacológico , Neoplasias Cutâneas/metabolismo , Resultado do Tratamento , Ácido Úrico/sangue , Ácido Úrico/metabolismo
12.
Biol Pharm Bull ; 41(4): 487-503, 2018 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-29332929

RESUMO

Detection of anomalous cells such as cancer cells from normal blood cells has the potential to contribute greatly to cancer diagnosis and therapy. Conventional methods for the detection of cancer cells are usually tedious and cumbersome. Herein, we report on the use of a particle size analyzer for the convenient size-based differentiation of cancer cells from normal cells. Measurements made using a particle size analyzer revealed that size parameters for cancer cells are significantly greater (e.g., inner diameter and width) than the corresponding values for normal cells (white blood cells (WBC), lymphocytes and splenocytes), with no significant difference in shape parameters (e.g., circularity and convexity). The inner diameter of many cancer cell lines is greater than 10 µm, in contrast to normal cells. For the detection of WBC having similar size to that of cancer cells, we developed a PC software "Cancer Cell Finder" that differentiates them from cancer cells based on brightness stationary points on a cell surface. Furthermore, the aforementioned method was validated for cancer cell/clusters detection in spiked mouse blood samples (a B16 melanoma mouse xenograft model) and circulating tumor cell cluster-like particles in the cat and dog (diagnosed with cancer) blood samples. These results provide insights into the possible applicability of the use of a particle size analyzer in conjunction with PC software for the convenient detection of cancer cells in experimental and clinical samples for theranostics.


Assuntos
Neoplasias da Mama/patologia , Neoplasias Colorretais/patologia , Neoplasias Pulmonares/patologia , Melanoma Experimental/patologia , Neoplasias da Próstata/patologia , Animais , Neoplasias da Mama/sangue , Neoplasias da Mama/diagnóstico , Gatos , Linhagem Celular Tumoral , Forma Celular , Tamanho Celular , Neoplasias Colorretais/sangue , Neoplasias Colorretais/diagnóstico , Cães , Feminino , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Humanos , Leucócitos/citologia , Leucócitos/patologia , Neoplasias Pulmonares/sangue , Neoplasias Pulmonares/diagnóstico , Linfócitos/citologia , Linfócitos/patologia , Masculino , Melanoma Experimental/sangue , Melanoma Experimental/diagnóstico , Camundongos Endogâmicos C57BL , Neoplasias da Próstata/sangue , Neoplasias da Próstata/diagnóstico , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Software , Baço/citologia , Baço/patologia , Propriedades de Superfície , Nanomedicina Teranóstica/instrumentação , Nanomedicina Teranóstica/métodos
13.
J Cancer Res Ther ; 13(6): 956-960, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-29237958

RESUMO

OBJECTIVE: The antitumor effect of ciprofloxacin has been widely assessed in-vitro, and positive results have been reported. The aim of this study was to investigate the influence of ciprofloxacin treatment on the growth of B16F10 melanoma cells both in-vitro and in-vivo. MATERIALS AND METHODS: Groups of C57BL/6 female mice challenged with B16F10 melanoma cells were kept untreated or were treated with sterile water, intraperitoneal ciprofloxacin, or ciprofloxacin through drinking water for 10 days. The serum levels of vascular endothelial growth factor (VEGF) were measured by ELISA 1 and 3 h after the last dose of ciprofloxacin. Mice were monitored for an additional 10 days for survival assessment. Moreover, B16F10 melanoma cells were cultured in 24-well plates and exposed to different concentrations of ciprofloxacin (10-1000 µg/ml). Viability was determined, after 24 and 48 h, using trypan blue. RESULTS: The serum levels of VEGF significantly decreased in ciprofloxacin-treated mice when compared to the controls. None of the control mice survived beyond day 8, whereas 16.67% of those treated with ciprofloxacin survived up to 18 days. In addition, the viability of B16F10 melanoma cells, in-vitro, significantly decreased with increasing concentrations of ciprofloxacin after 24 and 48 h. CONCLUSION: Ciprofloxacin seems to exhibit antitumor activity both in-vivo and in-vitro. This effect might be explained by several mechanisms such as directly inducing cancer cell death or altering the immune response through the modification of the normal microbiota.


Assuntos
Proliferação de Células/efeitos dos fármacos , Ciprofloxacina/administração & dosagem , Melanoma Experimental/tratamento farmacológico , Fator A de Crescimento do Endotélio Vascular/sangue , Animais , Linhagem Celular Tumoral , Ciprofloxacina/farmacocinética , Humanos , Melanoma Experimental/sangue , Melanoma Experimental/patologia , Camundongos
14.
Sci Rep ; 7(1): 16993, 2017 12 05.
Artigo em Inglês | MEDLINE | ID: mdl-29209060

RESUMO

This study presents, attenuated total reflection Fourier transforms infrared spectroscopy of dried serum samples in an effort to assess biochemical changes induced by non-Hodgkin's lymphoma and subcutaneous melanoma. An EL4 mouse model of non-Hodgkin lymphoma and a B16 mouse model of subcutaneous melanoma are used to extract a snapshot of tumor-associated alteration in the serum. The study of both cancer-bearing mouse models in wild types and their corresponding control types, emphasizes the diagnostic potential of this approach as a screening technique for non-Hodgkin lymphoma and melanoma skin cancer. Infrared absorbance values of the different spectral bands, hierarchical clustering and integral values of the component bands by curve fitting, show statistically significant differences (student's t-test, two-tailed unequal variance p-value < 0.05) between spectra representing healthy and tumorous mouse. This technique may thus be useful for having individualized route maps for rapid evaluation of lymphoma and melanoma status and associated therapeutic modalities.


Assuntos
Biomarcadores Tumorais/sangue , Linfoma/sangue , Linfoma/diagnóstico , Melanoma Experimental/sangue , Melanoma Experimental/diagnóstico , Espectroscopia de Infravermelho com Transformada de Fourier/métodos , Animais , Apoptose , Proliferação de Células , Diagnóstico Diferencial , Camundongos , Células Tumorais Cultivadas
15.
Methods Mol Biol ; 1634: 247-262, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28819857

RESUMO

The quantification of circulating tumor cells (CTCs) has been considered a potentially powerful tool in cancer diagnosis and prognosis, as CTCs have been shown to appear very early in cancer development. Great efforts have been made to develop methods that were less invasive and more sensitive to detect CTCs earlier. There is growing evidence that CTC clusters have greater metastatic potential than single CTCs. Therefore, the detection of CTC clusters is also important. This chapter is aimed to introduce a noninvasive technique for CTCs detection named in vivo flow cytometry (IVFC), which has been demonstrated to be capable of monitoring CTCs dynamics continuously. Furthermore, IVFC could be helpful for CTC cluster enumeration.


Assuntos
Citometria de Fluxo/métodos , Neoplasias Hepáticas/diagnóstico , Melanoma Experimental/diagnóstico , Células Neoplásicas Circulantes/patologia , Técnicas Fotoacústicas/instrumentação , Animais , Vasos Sanguíneos/metabolismo , Vasos Sanguíneos/ultraestrutura , Contagem de Células/instrumentação , Contagem de Células/métodos , Linhagem Celular Tumoral , Orelha/irrigação sanguínea , Citometria de Fluxo/instrumentação , Genes Reporter , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Humanos , Neoplasias Hepáticas/sangue , Neoplasias Hepáticas/patologia , Melanoma Experimental/sangue , Melanoma Experimental/patologia , Camundongos , Metástase Neoplásica , Células Neoplásicas Circulantes/metabolismo , Transfecção
16.
J Pharm Sci ; 106(8): 2037-2045, 2017 08.
Artigo em Inglês | MEDLINE | ID: mdl-28456734

RESUMO

Toll-like receptor (TLR) agonists TLR 7/8, MEDI9197, is a imidazoquinoline analogue that can be used for cancer immunotherapy based on its efficacy toward a variety of tumors. Systemic administration of TLR agonists results in stimulation of the immune system throughout the entire body causing undesirable side effects. To minimize these adverse events, local administration of TLR agonists including intratumoral (IT) delivery has been introduced. Here, a poloxamer 407 thermogel formulation for IT delivery of a TLR 7/8 dual agonist, MEDI9197, is described in which the combination of the aqueous thermogel and the ethanolic TLR 7/8 dual agonist, MEDI9197, solution leads to precipitated drug particles within the gel. The in vitro release profile showed an initial burst followed by sustained release. A B16-OVA mouse tumor model was used to assess the in vivo pharmacokinetics, efficacy, and systemic cytokine and chemokine (cytokine) production of the poloxamer 407-based thermogel formulation. The pharmacokinetic evaluation showed that the agonist level within the tumor was reduced by ∼70% over 14 days while serum agonist levels indicated an initial burst at the 6-h time point followed by a drop in serum drug levels over the 14 days of the experiment. The tumor growth inhibition, survival, and serum cytokines for post-IT injection of the poloxamer 407 formulation showed that it slowly released TLR 7/8 agonist, MEDI9197, resulting in more efficacious tumor growth inhibition compared with control groups. In addition, the cytokine levels in circulation indicated that a dose increase led to a decrease in the serum inflammatory and interferon-inducible cytokines levels. This observation could be due to a reduction of drug diffusion and escape from the tumor site due to the precipitation of the drug inside the tumor leading to sustained release. IT delivery of TLR 7/8 dual agonist, MEDI9197, via a thermosensitive gel-based formulation was efficacious and could offer an alternate method of local drug delivery.


Assuntos
Antineoplásicos/administração & dosagem , Preparações de Ação Retardada/química , Compostos Heterocíclicos com 3 Anéis/administração & dosagem , Melanoma Experimental/tratamento farmacológico , Poloxâmero/química , Ácidos Esteáricos/administração & dosagem , Receptor 7 Toll-Like/agonistas , Receptor 8 Toll-Like/agonistas , Animais , Antineoplásicos/farmacocinética , Antineoplásicos/uso terapêutico , Citocinas/sangue , Citocinas/imunologia , Sistemas de Liberação de Medicamentos , Feminino , Géis/química , Compostos Heterocíclicos com 3 Anéis/farmacocinética , Compostos Heterocíclicos com 3 Anéis/uso terapêutico , Melanoma Experimental/sangue , Melanoma Experimental/imunologia , Melanoma Experimental/patologia , Camundongos , Camundongos Endogâmicos C57BL , Ácidos Esteáricos/farmacocinética , Ácidos Esteáricos/uso terapêutico , Temperatura , Receptor 7 Toll-Like/imunologia , Receptor 8 Toll-Like/imunologia
17.
Drug Discov Ther ; 10(6): 314-322, 2017 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-27746419

RESUMO

We previously demonstrated that the oncogenic kinase PAK4, which both melanomas and normal melanocytes express at a very high level, is essential for their melanogenesis. In the present study, using the highly sensitive "Macaroni-Western" (IP-ATP-Glo) kinase assay, we investigated the melanogenic potential of another oncogenic kinase PAK1, which melanoma (B16F10) cells express only at a very minute level. After transfecting melanoma cells with PAK1-shRNA for silencing PAK1 gene, melanin content, tyrosinase activity, and kinase activity of PAK1 were compared between the wild-type and transfectants. We found that (i) PAK1 is significantly activated by melanogenic hormones such as IBMX (3-isobutyl-1-methyl xanthine) and α-MSH (melanocyte-stimulating hormone), (ii) silencing the endogenous PAK1 gene in melanoma cells through PAK1-specific shRNA reduces both melanin content and tyrosinase activity in the presence of both serum and melanogenic hormones to the basal level, (iii) the exogenously added wild-type PAK1 in the melanoma cells boosts the α-MSH-inducible melanin level by several folds without affecting the basal, and (iv) α-MSH/IBMX-induced melanogenesis hardly takes place in the absence of either serum or PAK1, clearly indicating that PAK1 is essential mainly for serum- and α-MSH/IBMX-dependent melanogenesis, but not the basal, in melanoma cells. The outcome of this study might provide the first scientific basis for explaining why a wide variety of herbal PAK1-blockers such as CAPE (caffeic acid phenethyl ester), curcumin and shikonin in cosmetics are useful for skin-whitening.


Assuntos
1-Metil-3-Isobutilxantina/farmacologia , Hormônios Estimuladores de Melanócitos/farmacologia , Melanoma Experimental/metabolismo , Fator de Crescimento Derivado de Plaquetas/metabolismo , Quinases Ativadas por p21/genética , Animais , Linhagem Celular Tumoral , Regulação para Baixo , Regulação Neoplásica da Expressão Gênica , Melaninas/metabolismo , Melanoma Experimental/sangue , Melanoma Experimental/genética , Camundongos , Modelos Biológicos , Interferência de RNA , Quinases Ativadas por p21/metabolismo
19.
Sci Rep ; 6: 29914, 2016 07 18.
Artigo em Inglês | MEDLINE | ID: mdl-27426915

RESUMO

Hypoxia is a hallmark of cancer that is strongly associated with invasion, metastasis, resistance to therapy and poor clinical outcome. Tumour hypoxia affects immune responses and promotes the accumulation of macrophages in the tumour microenvironment. However, the signals linking tumour hypoxia to tumour-associated macrophage recruitment and tumour promotion are incompletely understood. Here we show that the damage-associated molecular pattern High-Mobility Group Box 1 protein (HMGB1) is released by melanoma tumour cells as a consequence of hypoxia and promotes M2-like tumour-associated macrophage accumulation and an IL-10 rich milieu within the tumour. Furthermore, we demonstrate that HMGB1 drives IL-10 production in M2-like macrophages by selectively signalling through the Receptor for Advanced Glycation End products (RAGE). Finally, we show that HMGB1 has an important role in murine B16 melanoma growth and metastasis, whereas in humans its serum concentration is significantly increased in metastatic melanoma. Collectively, our findings identify a mechanism by which hypoxia affects tumour growth and metastasis in melanoma and depict HMGB1 as a potential therapeutic target.


Assuntos
Macrófagos/metabolismo , Melanoma Experimental/metabolismo , Melanoma Experimental/patologia , Hipóxia Tumoral , Animais , Proliferação de Células , Feminino , Proteína HMGB1/sangue , Proteína HMGB1/metabolismo , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Interleucina-10/metabolismo , Macrófagos/patologia , Masculino , Melanoma Experimental/sangue , Camundongos Endogâmicos C57BL , Fenótipo , Receptor para Produtos Finais de Glicação Avançada/metabolismo , Neoplasias Cutâneas/sangue , Neoplasias Cutâneas/metabolismo , Neoplasias Cutâneas/patologia
20.
Int J Cancer ; 139(10): 2270-6, 2016 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-27459381

RESUMO

Sunitinib and sorafenib are broad-spectrum tyrosine kinase inhibitors (TKI) targeting, for example, VEGF1-3, PDGFRb, RET, FLT3, CD117 (c-KIT) and CSF-1R cell membrane receptors thus suppressing tumor angiogenesis and cancer cell growth. Recently it has been suggested that the kinases targeted by Sunitinib and/or Sorafenib regulate leukocyte transmigration, which might in part be responsible for the often-observed reduction in tumor-associated myeloid derived suppressor cells and regulatory T cells. The aim of the current study is to determine whether sunitinib or sorafenib inhibit leukocyte extravasation. Sunitinib, sorafenib, or vehicle treated animals did not show any difference in leukocyte trafficking either in peritonitis or in vivo homing experiments, although sunitinib treatment effectively inhibited growth of B16 melanoma tumors in WT, SCID and SCID beige mice. Inhibition of tumor growth was associated with an increased number of infiltrating CD11b+ cells in the tumor, while the numbers of CD8, Gr-1 and F4/80 expressing cells were unchanged. In conclusion, the findings suggest that despite multiple targets with a potential role in leukocyte extravasation, neither sunitinib nor sorafenib effectively inhibits this process in vivo. Thus, the observed specific effect on CD11b cells among tumor infiltrating leukocytes is most likely an indirect effect.


Assuntos
Indóis/farmacologia , Leucócitos/efeitos dos fármacos , Niacinamida/análogos & derivados , Compostos de Fenilureia/farmacologia , Inibidores de Proteínas Quinases/farmacologia , Pirróis/farmacologia , Animais , Movimento Celular/efeitos dos fármacos , Feminino , Leucócitos/enzimologia , Leucócitos/imunologia , Leucócitos/patologia , Melanoma Experimental/sangue , Melanoma Experimental/tratamento farmacológico , Melanoma Experimental/imunologia , Melanoma Experimental/patologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos SCID , Niacinamida/farmacologia , Sorafenibe , Sunitinibe , Linfócitos T Reguladores/efeitos dos fármacos , Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/patologia , Migração Transendotelial e Transepitelial/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...