Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 522
Filtrar
1.
Adv Sci (Weinh) ; 8(15): 2002020, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-34386315

RESUMO

Biomimetic strategies are useful for designing potent vaccines. Decorating a nanoparticulate adjuvant with cell membrane fragments as the antigen-presenting source exemplifies, such as a promising strategy. For translation, a standardizable, consistent, and scalable approach for coating nanoadjuvant with the cell membrane is important. Here a turbulent mixing and self-assembly method called flash nanocomplexation (FNC) for producing cell membrane-coated nanovaccines in a scalable manner is demonstrated. The broad applicability of this FNC technique compared with bulk-sonication by using ten different core materials and multiple cell membrane types is shown. FNC-produced biomimetic nanoparticles have promising colloidal stability and narrow particle polydispersity, indicating an equal or more homogeneous coating compared to the bulk-sonication method. The potency of a nanovaccine comprised of B16-F10 cancer cell membrane decorating mesoporous silica nanoparticles loaded with the adjuvant CpG is then demonstrated. The FNC-fabricated nanovaccines when combined with anti-CTLA-4 show potency in lymph node targeting, DC antigen presentation, and T cell immune activation, leading to prophylactic and therapeutic efficacy in a melanoma mouse model. This study advances the design of a biomimetic nanovaccine enabled by a robust and versatile nanomanufacturing technique.


Assuntos
Antígeno CTLA-4/imunologia , Vacinas Anticâncer/uso terapêutico , Melanoma Experimental/prevenção & controle , Nanopartículas/uso terapêutico , Adjuvantes Imunológicos/uso terapêutico , Animais , Apresentação de Antígeno/efeitos dos fármacos , Biomimética , Antígeno CTLA-4/antagonistas & inibidores , Vacinas Anticâncer/imunologia , Células Dendríticas/efeitos dos fármacos , Células Dendríticas/imunologia , Humanos , Linfonodos/efeitos dos fármacos , Linfonodos/imunologia , Melanoma Experimental/imunologia , Camundongos , Linfócitos T/imunologia
2.
Neoplasia ; 23(9): 837-850, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34298233

RESUMO

Understanding how the extracellular matrix affects cancer development constitutes an emerging research field. Fibronectin and collagen are two intriguing matrix components found in cancer. Large concentrations of fibronectin or collagen type I have been implicated in poor prognosis in patients. In a mouse model, we had shown that genetically decreasing circulating fibronectin resulted in smaller tumors. We therefore aimed to manipulate fibronectin pharmacologically and determine how cancer development is affected. Deletion of fibronectin in human breast cancer cells (MDA-MB-231) using shRNA (knockdown: Kd) improved survival and diminished tumor burden in a model of metastatic lesions and in a model of local growth. Based on these findings, it seemed reasonable to attempt to prevent fibronectin accumulation using a bacterial derived peptide called pUR4. Treatment with this peptide for 10 days in the breast cancer local growth model or for 5 days in a melanoma skin cancer model (B16) was associated with a significant suppression of cancer growth. Treatment aimed at inhibiting collagen type I accumulation without interfering with fibronectin could not affect any changes in vivo. In the absence of fibronectin, diminished cancer progression was due to inhibition of proliferation, even though changes in blood vessels were also detected. Decreased proliferation could be attributed to decreased ERK phosphorylation and diminished YAP expression. In summary, manipulating fibronectin diminishes cancer progression, mostly by suppressing cell proliferation. This suggests that matrix modulation could be used as an adjuvant to conventional therapy as long as a decrease in fibronectin is obtained.


Assuntos
Fibronectinas/antagonistas & inibidores , Fibronectinas/metabolismo , Neoplasias/metabolismo , Neoplasias/prevenção & controle , Sequência de Aminoácidos , Animais , Fibronectinas/genética , Humanos , Melanoma Experimental/genética , Melanoma Experimental/metabolismo , Melanoma Experimental/patologia , Melanoma Experimental/prevenção & controle , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Nus , Células NIH 3T3 , Neoplasias/genética , Neoplasias/patologia , Fragmentos de Peptídeos/administração & dosagem , Fragmentos de Peptídeos/genética , Carga Tumoral/efeitos dos fármacos , Carga Tumoral/fisiologia , Ensaios Antitumorais Modelo de Xenoenxerto/métodos
3.
Dokl Biochem Biophys ; 498(1): 199-202, 2021 May.
Artigo em Inglês | MEDLINE | ID: mdl-34189650

RESUMO

Two monoclonal antibodies recognizing non-overlapping epitopes of the PRAME protein were injected into immunocompetent mice to study their influence on the growth of subcutaneous tumor nodes. The B16F10 murine melanoma line, either expressing human PRAME protein or bearing only a vector without PRAME gene, were used as transplants. Each of the antibodies showed the ability to suppress tumor growth of a PRAME-expressing tumour, but not a tumor without PRAME.


Assuntos
Anticorpos Monoclonais/farmacologia , Antígenos de Neoplasias/imunologia , Epitopos/imunologia , Melanoma Experimental/prevenção & controle , Animais , Anticorpos Monoclonais/administração & dosagem , Feminino , Melanoma Experimental/etiologia , Melanoma Experimental/metabolismo , Melanoma Experimental/patologia , Camundongos , Camundongos Endogâmicos C57BL
4.
Radiat Res ; 195(6): 522-540, 2021 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-33826741

RESUMO

Brain metastases develop in over 60% of advanced melanoma patients and negatively impact quality of life and prognosis. In a murine melanoma model, we previously showed that an in situ vaccination (ISV) regimen, combining radiation treatment and intratumoral (IT) injection of immunocytokine (IC: anti-GD2 antibody fused to IL2), along with the immune checkpoint inhibitor anti-CTLA-4, robustly eliminates peripheral flank tumors but only has modest effects on co-occurring intracranial tumors. In this study, we investigated the ability of low-dose radiation to the brain to potentiate anti-tumor immunity against a brain tumor when combined with ISV + anti-CTLA-4. B78 (GD2+, immunologically "cold") melanoma tumor cells were implanted into the flank and the right striatum of the brain in C57BL/6 mice. Flank tumors (50-150 mm3) were treated following a previously optimized ISV regimen [radiation (12 Gy × 1, treatment day 1), IT-IC (50 µg daily, treatment days 6-10), and anti-CTLA-4 (100 µg, treatment days 3, 6, 9)]. Mice that additionally received whole-brain radiation treatment (WBRT, 4 Gy × 1) on day 15 demonstrated significantly increased survival compared to animals that received ISV + anti-CTLA-4 alone, WBRT alone or no treatment (control) (P < 0.001, log-rank test). Timing of WBRT was critical, as WBRT administration on day 1 did not significantly enhance survival compared to ISV + anti-CTLA-4, suggesting that the effect of WBRT on survival might be mediated through immune modulation and not just direct tumor cell cytotoxicity. Modest increases in T cells (CD8+ and CD4+) and monocytes/macrophages (F4/80+) but no changes in FOXP3+ regulatory T cells (Tregs), were observed in brain melanoma tumors with addition of WBRT (on day 15) to ISV + anti-CTLA-4. Cytokine multiplex immunoassay revealed distinct changes in both intracranial melanoma and contralateral normal brain with addition of WBRT (day 15) to ISV + anti-CTLA-4, with notable significant changes in pro-inflammatory (e.g., IFNγ, TNFα and LIX/CXCL5) and suppressive (e.g., IL10, IL13) cytokines as well as chemokines (e.g., IP-10/CXCL10 and MIG/CXCL9). We tested the ability of the alkylphosphocholine analog, NM600, to deliver immunomodulatory radiation to melanoma brain tumors as a targeted radionuclide therapy (TRT). Yttrium-86 (86Y) chelated to NM600 was delivered intravenously by tail vein to mice harboring flank and brain melanoma tumors, and PET imaging demonstrated specific accumulation up to 72 h at each tumor site (∼12:1 brain tumor/brain and ∼8:1 flank tumor/muscle). When NM600 was chelated to therapeutic ß-particle-emitting 90Y and administered on treatment day 13, T-cell infiltration and cytokine profiles were altered in melanoma brain tumor, like that observed for WBRT. Overall, our results demonstrate that addition of low-dose radiation, timed appropriately with ISV administration to tumors outside the brain, significantly increases survival in animals co-harboring melanoma brain tumors. This observation has potentially important translational implications as a treatment strategy for increasing the response of tumors in the brain to systemically administered immunotherapies.


Assuntos
Neoplasias Encefálicas/imunologia , Imunidade/efeitos da radiação , Melanoma Experimental/imunologia , Vacinação , Animais , Neoplasias Encefálicas/prevenção & controle , Linhagem Celular Tumoral , Relação Dose-Resposta à Radiação , Inibidores de Checkpoint Imunológico/farmacologia , Imunidade/efeitos dos fármacos , Melanoma Experimental/prevenção & controle , Camundongos , Camundongos Endogâmicos C57BL , Proteína Tumoral 1 Controlada por Tradução
5.
J Biol Chem ; 296: 100523, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33711340

RESUMO

The Stimulator of Interferon Genes (STING) pathway is implicated in the innate immune response and is important in both oncogenesis and cancer treatment. Specifically, activation of the cytosolic DNA sensor STING in antigen-presenting cells (APCs) induces a type I interferon response and cytokine production that facilitates antitumor immune therapy. However, use of STING agonists (STINGa) as a cancer therapeutic has been limited by unfavorable pharmacological properties and targeting inefficiency due to rapid clearance and limited uptake into the cytosol. Exosomes, a class of extracellular vesicles shed by all cells are under consideration for their use as effective carriers of drugs owing to their innate ability to be taken up by cells and their biocompatibility for optimal drug biodistribution. Therefore, we engineered exosomes to deliver the STING agonist cyclic GMP-AMP (iExoSTINGa), to exploit their favorable pharmacokinetics and pharmacodynamics. Selective targeting of the STING pathway in APCs with iExoSTINGa was associated with superior potency compared with STINGa alone in suppressing B16F10 tumor growth. Moreover, iExoSTINGa showed superior uptake of STINGa into dendritic cells compared with STINGa alone, which led to increased accumulation of activated CD8+ T-cells and an antitumor immune response. Our study highlights the potential of exosomes in general, and iExoSTINGa specifically, in enhancing cancer therapy outcomes.


Assuntos
Células Apresentadoras de Antígenos/imunologia , Linfócitos T CD8-Positivos/imunologia , Exossomos/metabolismo , Imunidade Inata/imunologia , Melanoma Experimental/imunologia , Proteínas de Membrana/agonistas , Nucleotídeos Cíclicos/farmacologia , Animais , Células Apresentadoras de Antígenos/efeitos dos fármacos , Linfócitos T CD8-Positivos/efeitos dos fármacos , Feminino , Células HEK293 , Humanos , Imunidade Inata/efeitos dos fármacos , Melanoma Experimental/metabolismo , Melanoma Experimental/patologia , Melanoma Experimental/prevenção & controle , Camundongos , Camundongos Endogâmicos C57BL , Transdução de Sinais
6.
Nat Cell Biol ; 23(1): 75-86, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33420490

RESUMO

Nutrient availability is central for T-cell functions and immune responses. Here we report that CD8+ T-cell activation and anti-tumour responses are strongly potentiated by the non-essential amino acid Asn. Increased Asn levels enhance CD8+ T-cell activation and effector functions against tumour cells in vitro and in vivo. Conversely, restriction of dietary Asn, ASNase administration or inhibition of the Asn transporter SLC1A5 impairs the activity and responses of CD8+ T cells. Mechanistically, Asn does not directly alter cellular metabolic fluxes; it instead binds the SRC-family protein tyrosine kinase LCK and orchestrates LCK phosphorylation at Tyr 394 and 505, thereby leading to enhanced LCK activity and T-cell-receptor signalling. Thus, our findings reveal a critical and metabolism-independent role for Asn in the direct modulation of the adaptive immune response by controlling T-cell activation and efficacy, and further uncover that LCK is a natural Asn sensor signalling Asn sufficiency to T-cell functions.


Assuntos
Asparagina/farmacologia , Linfócitos T CD8-Positivos/imunologia , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Neoplasias Pulmonares/prevenção & controle , Ativação Linfocitária , Proteína Tirosina Quinase p56(lck) Linfócito-Específica/imunologia , Melanoma Experimental/prevenção & controle , Animais , Linfócitos T CD8-Positivos/efeitos dos fármacos , Feminino , Humanos , Neoplasias Pulmonares/imunologia , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/secundário , Masculino , Melanoma Experimental/imunologia , Melanoma Experimental/metabolismo , Melanoma Experimental/patologia , Camundongos , Camundongos Endogâmicos C57BL , Fosforilação , Transdução de Sinais , Células Tumorais Cultivadas , Tirosina/metabolismo , Quinases da Família src/metabolismo
7.
Immunotherapy ; 13(4): 309-326, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33397152

RESUMO

Aim: Whole dead tumor cells can be used as antigen source and the induction of protective immune response could be enhanced by damage-associated molecular patterns. Materials & methods: We generated whole dead tumor cells called B16-immunogenic cell bodies (ICBs) from B16 melanoma cells by nutrient starvation and evaluated the in vivo antitumor effect of B16-ICBs plus ATP and polymyxin B (PMB). Results: The subcutaneous immunization with B16-ICBs + PMB + ATP a 50% of tumor-free animals and induced a significant delay in tumor growth in a prophylactic approach. These results correlated with maturation of bone marrow-derived dendritic cells and activation of T CD8+ lymphocytes in vitro. Conclusion: Altogether, ICB + ATP + PMB is efficient in inducing the antitumor efficacy of the whole dead tumor cells vaccine.


Assuntos
Trifosfato de Adenosina/imunologia , Vacinas Anticâncer/imunologia , Melanoma Experimental/imunologia , Polimixina B/imunologia , Trifosfato de Adenosina/administração & dosagem , Alarminas/administração & dosagem , Alarminas/imunologia , Animais , Apresentação de Antígeno , Antígenos de Neoplasias/imunologia , Antígenos CD40/metabolismo , Vacinas Anticâncer/administração & dosagem , Citocinas/metabolismo , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Imunização , Melanoma Experimental/patologia , Melanoma Experimental/prevenção & controle , Camundongos , Camundongos Endogâmicos C57BL , Fagocitose , Polimixina B/administração & dosagem , Baço/imunologia , Células Tumorais Cultivadas
9.
Anticancer Agents Med Chem ; 21(6): 738-746, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-32723258

RESUMO

BACKGROUND: The antigen HCA587 (also known as MAGE-C2), which is considered a cancer-testis antigen, exhibits upregulated expression in a wide range of malignant tumors with unique immunological properties, and may thus serve as a promising target for tumor immunotherapy. OBJECTIVE: The study aimed to explore the antitumor effect of the HCA587 protein vaccine and the response of humoral and cell-mediated immunity. METHODS: The HCA587 protein vaccine was formulated with adjuvants CpG and ISCOM. B16 melanoma cells were subcutaneously inoculated to C57BL/6 mice, followed by treatment with HCA587 protein vaccine subcutaneously. Mouse survival was monitored daily, and tumor volume was measured every 2 to 3 days. The tumor sizes, survival time and immune cells in tumor tissues were detected. And the vital immune cell subset and effector molecules were explored. RESULTS: After treatment with HCA587 protein vaccine, the vaccination elicited significant immune responses, which delayed tumor growth and improved animal survival. The vaccination increased the proportion of CD4+ T cells expressing IFN-γ and granzyme B in tumor tissues. The depletion of CD4+T cells resulted in an almost complete abrogation of the antitumor effect of the vaccination, suggesting that the antitumor efficacy was mediated by CD4+ T cells. In addition, knockout of IFN-γ resulted in a decrease in granzyme B levels, which were secreted by CD4+ T cells, and the antitumor effect was also significantly attenuated. CONCLUSION: The HCA587 protein vaccine may increase the levels of granzyme B expressed by CD4+ T cells, and this increase is dependent on IFN-γ, and the vaccine resulted in a specific tumor immune response and subsequent eradication of the tumor.


Assuntos
Antígenos de Neoplasias/imunologia , Linfócitos T CD4-Positivos/imunologia , Vacinas Anticâncer/imunologia , Granzimas/imunologia , Melanoma Experimental/prevenção & controle , Proteínas de Neoplasias/imunologia , Adjuvantes Imunológicos , Animais , Composição de Medicamentos , Regulação da Expressão Gênica/imunologia , Granzimas/genética , Humanos , Imunidade , Imunoterapia , Interferon gama/genética , Interferon gama/imunologia , Masculino , Melanoma Experimental/genética , Melanoma Experimental/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Neoplasias Experimentais
10.
J Virol ; 95(3)2021 01 13.
Artigo em Inglês | MEDLINE | ID: mdl-33177208

RESUMO

Oncolytic virotherapy (OVT) is now understood to be an immunotherapy that uses viral infection to liberate tumor antigens in an immunogenic context to promote the development of antitumor immune responses. The only currently FDA-approved oncolytic virotherapy, T-Vec, is a modified type 1 herpes simplex virus (HSV-1). While T-Vec is associated with limited response rates, its modest efficacy supports the continued development of novel OVT viruses. Herein, we test the efficacy of a recombinant HSV-1, VC2, as an OVT in a syngeneic B16F10-derived mouse model of melanoma. VC2 possesses mutations that block its ability to enter neurons via axonal termini. This greatly enhances its safety profile by precluding the ability of the virus to establish latent infection. VC2 has been shown to be a safe, effective vaccine against both HSV-1 and HSV-2 infection in mice, guinea pigs, and nonhuman primates. We found that VC2 slows tumor growth rates and that VC2 treatment significantly enhances survival of tumor-engrafted, VC2-treated mice over control treatments. VC2-treated mice that survived initial tumor engraftment were resistant to a second engraftment as well as colonization of lungs by intravenous introduction of tumor cells. We found that VC2 treatment induced substantial increases in intratumoral T cells and a decrease in immunosuppressive regulatory T cells. This immunity was critically dependent on CD8+ T cells and less dependent on CD4+ T cells. Our data provide significant support for the continued development of VC2 as an OVT for the treatment of human and animal cancers.IMPORTANCE Current oncolytic virotherapies possess limited response rates. However, when certain patient selection criteria are used, oncolytic virotherapy response rates have been shown to increase. This, in addition to the increased response rates of oncolytic virotherapy in combination with other immunotherapies, suggests that oncolytic viruses possess significant therapeutic potential for the treatment of cancer. As such, it is important to continue to develop novel oncolytic viruses as well as support basic research into their mechanisms of efficacy. Our data demonstrate significant clinical potential for VC2, a novel type 1 oncolytic herpes simplex virus. Additionally, due to the high rates of survival and the dependence on CD8+ T cells for efficacy, our model will enable study of the immunological correlates of protection for VC2 oncolytic virotherapy and oncolytic virotherapy in general. Understanding the mechanisms of efficacious oncolytic virotherapy will inform the rational design of improved oncolytic virotherapies.


Assuntos
Herpesvirus Humano 1/genética , Neoplasias Pulmonares/prevenção & controle , Melanoma Experimental/prevenção & controle , Terapia Viral Oncolítica/métodos , Linfócitos T Reguladores/imunologia , Animais , Modelos Animais de Doenças , Feminino , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/imunologia , Neoplasias Pulmonares/secundário , Melanoma Experimental/genética , Melanoma Experimental/imunologia , Melanoma Experimental/patologia , Camundongos , Camundongos Endogâmicos C57BL
11.
Nat Nanotechnol ; 15(12): 1043-1052, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-33139933

RESUMO

Cancer metastases and recurrence after surgical resection remain an important cause of treatment failure. Here we demonstrate a general strategy to fabricate personalized nanovaccines based on a cationic fluoropolymer for post-surgical cancer immunotherapy. Nanoparticles formed by mixing the fluoropolymer with a model antigen ovalbumin, induce dendritic cell maturation via the Toll-like receptor 4 (TLR4)-mediated signalling pathway, and promote antigen transportation into the cytosol of dendritic cells, which leads to an effective antigen cross-presentation. Such a nanovaccine inhibits established ovalbumin-expressing B16-OVA melanoma. More importantly, a mix of the fluoropolymer with cell membranes from resected autologous primary tumours synergizes with checkpoint blockade therapy to inhibit post-surgical tumour recurrence and metastases in two subcutaneous tumour models and an orthotopic breast cancer tumour. Furthermore, in the orthotopic tumour model, we observed a strong immune memory against tumour rechallenge. Our work offers a simple and general strategy for the preparation of personalized cancer vaccines to prevent post-operative cancer recurrence and metastasis.


Assuntos
Vacinas Anticâncer/uso terapêutico , Polímeros de Fluorcarboneto/uso terapêutico , Melanoma Experimental/prevenção & controle , Nanopartículas/uso terapêutico , Animais , Vacinas Anticâncer/química , Células Cultivadas , Feminino , Polímeros de Fluorcarboneto/química , Imunoterapia , Melanoma Experimental/imunologia , Melanoma Experimental/cirurgia , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Ovalbumina/química , Ovalbumina/uso terapêutico , Polietilenoimina/química , Polietilenoimina/uso terapêutico
12.
Carbohydr Polym ; 250: 116869, 2020 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-33049818

RESUMO

Melanoma is the most lethal form of skin cancer, with a worldwide increase in incidence. Despite the increased overall survival of metastatic melanoma patients given recent advances in targeted and immunotherapy, it still has a poor prognosis and available treatment options carry diverse severe side effects. Polysaccharides from seaweed have been shown to exert antitumor activities. Here we show in vitro and in vivo antitumor activities of a sulfated homogalactan (named 3G4S) from Codium isthmocladum seaweed in the B16-F10 murine melanoma cell line. 3G4S did not induce cytotoxicity or proliferation changes; however, it was able to reduce solid tumor growth and metastasis, while not inducing side effects in mice. B16-F10 cells traits related to the metastatic cascade were also impaired by 3G4S, reducing cell invasion, colony-forming capacity and membrane glycoconjugates. Therefore, 3G4S shows promising antitumor activities without the commonly associated drawbacks of cancer treatments and can be further explored.


Assuntos
Galactanos/farmacologia , Química Verde , Melanoma Experimental/prevenção & controle , Alga Marinha/química , Sulfatos/química , Animais , Apoptose , Proliferação de Células , Feminino , Humanos , Masculino , Melanoma Experimental/secundário , Camundongos , Camundongos Endogâmicos C57BL , Células Tumorais Cultivadas
13.
Mol Carcinog ; 59(11): 1292-1301, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32924161

RESUMO

Ultraviolet B (UVB) exposure is a core factor that leads to skin disease or carcinogenesis through the insufficient repair of DNA lesions. UVB-induced DNA lesions are mainly removed by the nucleotide excision repair (NER) mechanism. The expression of histone deacetylase 4 (HDAC4) is altered in the skin upon UVB exposure, indicating its possible implication in UVB-induced DNA lesions repair. Here, we investigated the role of HDAC4 in the NER removal of the main classes of UVB-induced DNA lesions consisting of cyclobutane pyrimidine dimers and pyrimidine (6-4) pyrimidone photoproducts (6-4PPs). We found that UVB irradiation increased HDAC4 expression at both the mRNA and protein levels. HDAC4 interacted with NER factor XPC, which played an important role in effectively removing the UVB-induced DNA lesions. This study provides an understanding of the HDAC4 function in DNA repair, which will allow the development of efficient strategies to protect the skin from UVR-induced diseases.


Assuntos
Dano ao DNA , Reparo do DNA , Histona Desacetilases/metabolismo , Melanoma Experimental/prevenção & controle , Substâncias Protetoras , Neoplasias Cutâneas/prevenção & controle , Raios Ultravioleta/efeitos adversos , Animais , Histona Desacetilases/genética , Melanoma Experimental/etiologia , Melanoma Experimental/patologia , Camundongos , Neoplasias Cutâneas/etiologia , Neoplasias Cutâneas/patologia , Células Tumorais Cultivadas
14.
Int J Mol Sci ; 21(16)2020 Aug 13.
Artigo em Inglês | MEDLINE | ID: mdl-32823603

RESUMO

We explored the effect of a recombinant mucin1-maltose-binding protein vaccine, including immunization cycles of recombinant mucin1-maltose-binding protein (MUC1-MBP) and CpG 2006 on T cell responses to human MUC1-overexpressing mouse melanoma B16 cells (B16-MUC1) melanoma in mice. We found that the vaccine had a significant antitumor effect, with the most obvious tumor-suppressive effect being observed in mice immunized five times. After more than five immunizations, the tumor inhibition rate decreased from 81.67% (five immunizations) to 43.67% (eight immunizations). To study the possible mechanism, Mucin-1(MUC1)-specific antibodies, IFN-γ secretion by lymphocytes, and cytotoxic T lymphocyte (CTL) cytotoxicity were measured by enzyme-linked immunosorbent assay (ELISA) and a real-time cell analyzer (RTCA). T cell subsets and immunosuppressive cells in the mouse spleen and tumor microenvironment were analyzed by FACS. These results showed that five immunizations activated MUC1-specific Th1 and CTL and reduced the ratio of myeloid-derived suppressor cells (MDSCs) and Th17 in mice more significantly than eight immunizations, indicating that excessive frequency of the immune cycle leads to the increased numbers of immunosuppressive cells and decreased numbers of immunostimulatory cells, thereby inhibiting antitumor immune activity. This data provide an experimental foundation for the clinical application of a recombinant MUC1-MBP vaccine.


Assuntos
Imunização , Proteínas Ligantes de Maltose/imunologia , Melanoma Experimental/imunologia , Mucina-1/imunologia , Linfócitos T/imunologia , Vacinas Sintéticas/imunologia , Animais , Proliferação de Células , Modelos Animais de Doenças , Feminino , Humanos , Melanoma Experimental/prevenção & controle , Camundongos Endogâmicos C57BL , Baço/patologia , Microambiente Tumoral , Regulação para Cima
15.
Biomolecules ; 10(8)2020 07 24.
Artigo em Inglês | MEDLINE | ID: mdl-32722030

RESUMO

Obesity is known to be associated with risk and aggressiveness of cancer. Melanoma, the most lethal type of skin cancer, is also closely related to the prevalence of obesity. In this study, we established a cancer-obesity comorbidity (COC) model to investigate the effects of vanillic acid (VA). After a five-week administration with a high-fat diet (HFD) to induce obesity, subcutaneous allograft of B16BL6 cells were followed, and VA was orally administrated for an additional two weeks. VA-fed mice showed significantly decreased body weight and white adipose tissue (WAT) weight, which were due to increased thermogenesis and AMPK activation in WATs. Growth of cancer was also suppressed. Mechanistic studies revealed increased apoptosis and autophagy markers by VA; however, caspase 3 was not involved. Since signal transducer and activator of transcription 3 (STAT3) is suggested as an important pathway linking obesity and cancer, we further investigated to find out if STAT3 phosphorylation was repressed by VA treatment, and this was again confirmed in a COC cell model of adipocyte conditioned medium-treated B16BL6 melanoma cells. Overall, our results show VA induces STAT3-mediated autophagy to inhibit cancer growth and thermogenesis to ameliorate obesity in COC. Based on these findings, we suggest VA as a candidate therapeutic agent for COC treatment.


Assuntos
Melanoma Experimental/prevenção & controle , Obesidade/prevenção & controle , Fator de Transcrição STAT3/metabolismo , Ácido Vanílico/farmacologia , Células 3T3-L1 , Adipócitos/citologia , Tecido Adiposo Branco/efeitos dos fármacos , Tecido Adiposo Branco/metabolismo , Tecido Adiposo Branco/patologia , Animais , Autofagia/efeitos dos fármacos , Peso Corporal/efeitos dos fármacos , Meios de Cultivo Condicionados/farmacologia , Dieta Hiperlipídica/efeitos adversos , Lipólise/efeitos dos fármacos , Masculino , Melanoma Experimental/complicações , Melanoma Experimental/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Obesidade/complicações , Obesidade/etiologia , Tamanho do Órgão/efeitos dos fármacos , Fosforilação/efeitos dos fármacos , Termogênese/efeitos dos fármacos
16.
Nutrients ; 12(7)2020 Jul 19.
Artigo em Inglês | MEDLINE | ID: mdl-32707654

RESUMO

Black cumin (Nigella sativa) seed extract has been shown to improve dermatological conditions, yet its beneficial effects for skin are not fully elucidated. Herein, Thymocid®, a chemically standardized black cumin seed extract, was investigated for its cosmeceutical potential including anti-aging properties associated with modulation of glycation, collagen cross-linking, and collagenase and elastase activities, as well as antimelanogenic effect in murine melanoma B16F10 cells. Thymocid® (50, 100, and 300 µg/mL) inhibited the formation of advanced glycation end-products (by 16.7-70.7%), collagen cross-linking (by 45.1-93.3%), collagenase activity (by 10.4-92.4%), and elastases activities (type I and III by 25.3-75.4% and 36.0-91.1%, respectively). In addition, Thymocid® (2.5-20 µg/mL) decreased melanin content in B16F10 cells by 42.5-61.6% and reduced cellular tyrosinase activity by 20.9% (at 20 µg/mL). Furthermore, Thymocid® (20 µg/mL for 72 h) markedly suppressed the mRNA expression levels of melanogenesis-related genes including microphthalmia-associated transcription factor (MITF), tyrosinase-related protein 1 (TYRP1), and TYRP2 to 78.9%, 0.3%, and 0.2%, respectively. Thymocid® (10 µg/mL) also suppressed the protein expression levels of MITF (by 15.2%) and TYRP1 (by 97.7%). Findings from this study support the anti-aging and antimelanogenic potential of Thymocid® as a bioactive cosmeceutical ingredient for skin care products.


Assuntos
Colágeno/metabolismo , Colagenases/metabolismo , Oxirredutases Intramoleculares/metabolismo , Melaninas/metabolismo , Melanoma Experimental/metabolismo , Melanoma Experimental/prevenção & controle , Glicoproteínas de Membrana/metabolismo , Fator de Transcrição Associado à Microftalmia/metabolismo , Nigella sativa/química , Oxirredutases/metabolismo , Elastase Pancreática/metabolismo , Fitoterapia , Extratos Vegetais/farmacologia , Sementes/química , Animais , Linhagem Celular Tumoral , Cosméticos , Produtos Finais de Glicação Avançada/metabolismo , Camundongos , Extratos Vegetais/uso terapêutico , Higiene da Pele
17.
Int Immunopharmacol ; 76: 105872, 2019 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-31499268

RESUMO

Melanoma cells are significantly resistance to the current treatments. Therefore, the best option for high-risk populations is prevention. Recently, many preventive cancer vaccines have been developed. In our previous study, several bioinformatic tools were employed for selection of the most immunodominant epitopes of acrosin binding protein (ACRBP) and synaptonemal complex protein 1 (SYCP1) antigens to design multiepitope DNA and peptide cancer vaccines. In the current study, the final construct of the multiepitope DNA vaccine was placed into a pcDNA3.1 vector and then, subcloned into a pET-28a (+) expression vector for transfecting BL21 E. coli strain. The recombinant multiepitope peptide vaccine, weighing 6.35 kDa, was purified by Fast protein liquid chromatography technique (FPLC) and detected by western blotting. Subsequently, C57BL/6 mice were immunized by a mixture of the peptide vaccine and incomplete Freund's adjuvant (IFA) (four vaccinations with one-week intervals). Two weeks after the last vaccination, the serum levels of the peptide-specific IgG total, IgG2a, and IgG1 were measured by enzyme-linked immunosorbent assays (ELISA). Also, the immunized mice splenocytes efficacy for producing interleukin-4 (IL-4) and interferon-γ (IFN-γ) after stimulation with the peptide vaccine was evaluated. At last, the prophylactic effect of the peptide vaccine immunization was evaluated in B16-F10 murine melanoma model. The peptide vaccine immunization caused a significant increase in the serum levels of IgG1, IgG2a, and IgG2a. Also, the immunized mice splenocytes exhibited significantly higher ability to produce IL-4 (10-fold) and IFN-γ (16-fold) after stimulation with the peptide vaccine, in comparison with the PBS and IFA groups. The peptide immunized mice exhibited 50.2% and 43% decrease in the mean tumors' volume in comparison with PBS and IFA groups. Also, the mean survival time for the peptide immunized mice was 33 ±â€¯1.3 days which was 5 and 6 days more than the PBS and IFA groups, respectively. The obtained results exhibit high efficacy of the designed multiepitope peptide vaccine for the immune system activation and anti-tumor prophylactic effects in the murine melanoma model.


Assuntos
Acrossomo/imunologia , Vacinas Anticâncer , Proteínas de Transporte/imunologia , Proteínas de Ligação a DNA/imunologia , Melanoma Experimental/prevenção & controle , Vacinas de DNA , Vacinas de Subunidades Antigênicas , Animais , Antígenos/genética , Antígenos/imunologia , Proteínas de Transporte/genética , Linhagem Celular Tumoral , Proteínas de Ligação a DNA/genética , Feminino , Epitopos Imunodominantes/imunologia , Imunoglobulina G/sangue , Linfócitos/efeitos dos fármacos , Linfócitos/imunologia , Melanoma Experimental/patologia , Camundongos Endogâmicos C57BL , Carga Tumoral
18.
Life Sci ; 233: 116711, 2019 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-31374233

RESUMO

AIMS: Insulin is a central peptide hormone required for carbohydrate metabolism; however, its role in diabetes-associated pulmonary disease is unknown. Here, we investigated the preventative effect of insulin against hyperglycemia-induced pulmonary vascular leakage and its molecular mechanism of action in the lungs of diabetic mice. MAIN METHODS: Vascular endothelial growth factor (VEGF) activated transglutaminase 2 (TGase2) by sequentially elevating intracellular Ca2+ and reactive oxygen species (ROS) levels in primary human pulmonary microvascular endothelial cells (HPMVECs). KEY FINDINGS: Insulin inhibited VEGF-induced TGase2 activation, but did not affect intracellular Ca2+ elevation and ROS generation. Insulin prevented VEGF-induced vascular leakage by inhibiting TGase2-mediated c-Src phosphorylation, disassembly of VE-cadherin and ß-catenin, and stress fiber formation. Insulin replacement therapy prevented hyperglycemia-induced TGase2 activation, but not ROS generation, in the lungs of diabetic mice. Insulin also prevented vascular leakage and cancer metastasis in the diabetic lung. Notably, vascular leakage was not detectable in the lungs of TGase2-null (Tgm2-/-) diabetic mice. SIGNIFICANCE: These findings demonstrate that insulin prevents hyperglycemia-induced pulmonary vascular leakage in diabetic mice by inhibiting VEGF-induced TGase2 activation rather than ROS generation.


Assuntos
Diabetes Mellitus Experimental/fisiopatologia , Proteínas de Ligação ao GTP/antagonistas & inibidores , Hemorragia/prevenção & controle , Hipoglicemiantes/farmacologia , Insulina/farmacologia , Pneumopatias/prevenção & controle , Transglutaminases/antagonistas & inibidores , Animais , Proteínas de Ligação ao GTP/fisiologia , Hemorragia/etiologia , Hemorragia/patologia , Humanos , Pneumopatias/etiologia , Pneumopatias/patologia , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/prevenção & controle , Neoplasias Pulmonares/secundário , Masculino , Melanoma Experimental/metabolismo , Melanoma Experimental/patologia , Melanoma Experimental/prevenção & controle , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteína 2 Glutamina gama-Glutamiltransferase , Transglutaminases/fisiologia , Células Tumorais Cultivadas
19.
Vaccine ; 37(35): 4947-4955, 2019 08 14.
Artigo em Inglês | MEDLINE | ID: mdl-31307876

RESUMO

A major obstacle to obtaining relevant results in cancer vaccination has been the lack of identification of immunogenic antigens. Dendritic cell (DC)-based cancer vaccines used preventively may afford protection against tumor inoculation, but the effect of antigen choice on anti-tumor protection is not clear. When using irradiated syngeneic tumor cells to load DCs, tumor self-antigens are provided, including tumor-associated antigens (TAAs) and neoantigens generated by tumor mutations. On the other hand, allogeneic tumor cells could only supply shared TAAs. To assess the advantages of each source in protective vaccination, we analyzed in C57BL/6 mice the effect of loading DCs with irradiated syngeneic B16-F1 or allogeneic Cloudman melanoma cells; both cell lines were characterized by whole exome sequencing and RNAseq. Tumor cell components from the two irradiated cell lines were efficiently internalized by DCs, and transported to MHC-class II positive tubulovesicular compartments (MIICs). DCs loaded with allogeneic irradiated Cloudman cells (DC-ApoNecALLO) induced a partially effective anti-melanoma protection, although Cloudman and B16-F1 cells share the expression of melanocyte differentiation antigens (MDAs), cancer-testis antigens (CTAs) and other TAAs. DCs loaded with syngeneic B16-F1 cells (DC-ApoNecSYN) established a more potent and long-lasting protection and induced a humoral anti-B16F1 response, thus suggesting that neoepitopes are needed for inducing long-lasting protection.


Assuntos
Células Alógenas/imunologia , Antígenos de Neoplasias/imunologia , Vacinas Anticâncer/imunologia , Células Dendríticas/imunologia , Melanoma Experimental/imunologia , Animais , Linhagem Celular Tumoral , Modelos Animais de Doenças , Imunidade Humoral , Masculino , Melanoma Experimental/prevenção & controle , Camundongos , Camundongos Endogâmicos C57BL , Organismos Livres de Patógenos Específicos , Vacinação
20.
Free Radic Res ; 53(4): 418-429, 2019 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-31020875

RESUMO

Melanoma is the most aggressive skin cancer due to a high propensity for metastasis, with a 10-year survival rate of less than 10%. The devastating clinical outcome and lack of effective preventative therapeutics for metastatic melanoma necessitate the development of new therapeutic strategies targeted to inhibit the regulatory circuits underlying the progression and metastasis of melanoma. Melanoma metastasis requires migration and invasion of the malignant tumour cells driven by proteolytic remodelling of the extracellular matrix (ECM) executed by matrix metalloproteinases (MMPs), particularly MMP-2 and MMP-9. Inhibiting components of these circuits defines new therapeutic opportunities for melanoma with metastatic malignancy. Oxalomalate (OMA) is a competitive inhibitor of NADP+-dependent isocitrate dehydrogenase (IDH), which plays an important role in cellular signalling pathways regulated by reactive oxygen species (ROS). In this study, we investigated the therapeutic role of OMA in metastatic melanoma and the associated underlying mechanism of action. We report that OMA-mediated inhibition of IDH enzymes suppresses metastatic melanoma through inhibition of invasive cell migration based on MMP-9-mediated proteolytic remodelling of the ECM. In particular, our study provides the mechanistic foundation that OMA reduces the expression and secretion of MMP-9 through LKB1-mediated PEA3 degradation via the ROS-dependent ATM-Chk2-p53 signalling axis, resulting from inhibition of IDH enzymes. These results provide evidence that OMA targeting of the stress response to ROS by IDH inhibition is a promising therapy for the treatment of metastatic melanoma.


Assuntos
Isocitrato Desidrogenase/metabolismo , Neoplasias Pulmonares/prevenção & controle , Melanoma Experimental/prevenção & controle , Oxalatos/farmacologia , Estresse Oxidativo/efeitos dos fármacos , Espécies Reativas de Oxigênio/metabolismo , Animais , Movimento Celular , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/secundário , Masculino , Melanoma Experimental/metabolismo , Melanoma Experimental/patologia , Camundongos , Camundongos Endogâmicos BALB C , Transdução de Sinais
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA