Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
1.
Sci Total Environ ; 899: 165607, 2023 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-37474070

RESUMO

Contamination of disposable medical masks has become a growing problem globally in the wake of Covid-19 due to their widespread use and improper disposal. Three different mask layers, namely the outer layer, the meltblown (MB) filler layer and the inner layers release three different types of microplastics, whose physical and chemical properties change after prolonged environmental weathering. In this study, physical and chemical changes of mask microplastics before and after aging were characterized by different characterization techniques. The toxic effect and mechanism of aged mask microplastics on Escherichia coli (E. coli) were studied by measuring the growth inhibition of mask microplastics, the change in ATPase activity, the change in malondialdehyde content and reactive oxygen species production, and the release of the chemical composition of exopolymeric substances (EPS). The microplastics of the aged MB filter layer had the most significant inhibitory effect on E. coli growth, reaching 19.2 % after 36 h. Also, under the influence of mask microplastics, ATPase activity of E. coli was inhibited and a large amount of EPS was released. The chemical composition of EPS has also changed. This study proposed the possible toxicity mechanism of mask microplastics and the self-protection mechanism of E. coli, and provided a reference for future research on the toxic effects of mask microplastics on environmental organisms.


Assuntos
COVID-19 , Microplásticos , Humanos , Idoso , Microplásticos/toxicidade , Plásticos/toxicidade , Escherichia coli , Polímeros , Adenosina Trifosfatases , Máscaras
2.
Ecotoxicol Environ Saf ; 227: 112903, 2021 Dec 20.
Artigo em Inglês | MEDLINE | ID: mdl-34673417

RESUMO

As a new pollutant, microplastics have increasingly drawn public attention to its toxic behavior in the environment. The aim was to investigate the effect of styrene-butadiene-rubber microplastics (mSBR) with different degrees of aging on petroleum hydrocarbon (PHC) degrading bacteria in an environment with simultaneously existing pollutants. A series of experiments were carried out to investigate the changes in the physical and chemical properties of mSBR with aging and to examine the influence of these changes on the inhibition of PHC-degrading bacteria by mSBR in the vicinity of coexisting pollutants. The results showed that in the early stage of ultraviolet aging (10d), the particle surface shows wrinkles, but the structure is intact. After reaching the late stage of aging (20d), nano-scale fragments were generated on the surface of mSBR, the average particle size decreased from 3.074 µm to 2.297 µm, and the zeta potential increased from - 25.1 mV to - 33.1 mV. The inhibitory effect of bacteria is greater. At the same time, these changes in the physicochemical properties increase the adsorption effect of Cd by 20%, and also improve the stability of mSBR in solution, whereby bacterial growth is inhibited by inhibiting the LPO activity and protein concentration of PHC degrading bacteria.


Assuntos
Poluição por Petróleo , Petróleo , Poluentes Químicos da Água , Bactérias , Biodegradação Ambiental , Butadienos/toxicidade , Elastômeros , Hidrocarbonetos , Microplásticos , Petróleo/toxicidade , Plásticos/toxicidade , Estirenos , Poluentes Químicos da Água/análise , Poluentes Químicos da Água/toxicidade
3.
J Immunother Cancer ; 9(1)2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33408093

RESUMO

BACKGROUND: Despite the remarkable benefits associated with the interventional treatment of melanomas (and other solid cancers) with immune checkpoint and Braf inhibitors (Brafi), most patients ultimately progress on therapy. The presence of multifocal/disseminated disease in patients increases their mortality risk. Hence, the development of novel strategies to effectively treat patients with melanomas that are resistant to anti-PD1 mAb (αPD1) and/or Brafi, particularly those with multifocal/disseminated disease remains a major unmet clinical need. METHODS: Mice developing induced/spontaneous BrafV600E/Pten-/- melanomas were treated by cutaneous immunization with a DNA vaccine encoding the melanoma-associated antigen TRP2, with Brafi or αPD1 alone, or with a combination of these treatments. Tumor progression, tumor-infiltration by CD4+ and CD8+ T cells, and the development of TRP2-specific CD8+ T cells were then monitored over time. RESULTS: Vaccination led to durable antitumor immunity against PD1/Brafi-resistant melanomas in both single lesion and multifocal disease models, and it sensitized PD1-resistant melanomas to salvage therapy with αPD1. The therapeutic efficacy of the vaccine was associated with host skin-resident cells, the induction of a systemic, broadly reactive IFNγ+CD8+ T cell repertoire, increased frequencies of CD8+ TIL and reduced levels of PD1hi/intCD8+ T cells. Extended survival was associated with improved TIL functionality, exemplified by the presence of enhanced levels of IFNγ+CD8+ TIL and IL2+CD4+ TIL. CONCLUSIONS: These data support the use of a novel genetic vaccine for the effective treatment of localized or multifocal melanoma refractory to conventional αPD1-based and/or Brafi-based (immune)therapy.


Assuntos
Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Inibidores de Checkpoint Imunológico/administração & dosagem , Oxirredutases Intramoleculares/administração & dosagem , Melanoma/tratamento farmacológico , Inibidores de Proteínas Quinases/administração & dosagem , Vacinas de DNA/administração & dosagem , Animais , Linfócitos T CD8-Positivos/metabolismo , Sinergismo Farmacológico , Feminino , Humanos , Inibidores de Checkpoint Imunológico/uso terapêutico , Imunização , Oxirredutases Intramoleculares/genética , Oxirredutases Intramoleculares/imunologia , Masculino , Melanoma/genética , Melanoma/imunologia , Camundongos , Mutação , PTEN Fosfo-Hidrolase/genética , Inibidores de Proteínas Quinases/farmacologia , Proteínas Proto-Oncogênicas B-raf/antagonistas & inibidores , Proteínas Proto-Oncogênicas B-raf/genética , Tamoxifeno/efeitos adversos , Tamoxifeno/análogos & derivados , Resultado do Tratamento , Vacinas de DNA/genética , Vacinas de DNA/imunologia , Ensaios Antitumorais Modelo de Xenoenxerto
4.
Cancer Res ; 79(10): 2736-2747, 2019 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-30914431

RESUMO

The tumor microenvironment has been compared with a nonhealing wound involving a complex interaction between multiple cell types. Schwann cells, the key regulators of peripheral nerve repair, have recently been shown to directly affect nonneural wound healing. Their role in cancer progression, however, has been largely limited to neuropathic pain and perineural invasion. In this study, we showed that melanoma activated otherwise dormant functions of Schwann cells aimed at nerve regeneration and wound healing. Such reprogramming of Schwann cells into repair-like cells occurred during the destruction and displacement of neurons as the tumor expanded and via direct signaling from melanoma cells to Schwann cells, resulting in activation of the nerve injury response. Melanoma-activated Schwann cells significantly altered the microenvironment through their modulation of the immune system and the extracellular matrix in a way that promoted melanoma growth in vitro and in vivo. Local inhibition of Schwann cell activity following cutaneous sensory nerve transection in melanoma orthotopic models significantly decreased the rate of tumor growth. Tumor-associated Schwann cells, therefore, can have a significant protumorigenic effect and may present a novel target for cancer therapy. SIGNIFICANCE: These findings reveal a role of the nerve injury response, particularly through functions of activated Schwann cells, in promoting melanoma growth.


Assuntos
Proliferação de Células/fisiologia , Melanoma/patologia , Células de Schwann/patologia , Transdução de Sinais/fisiologia , Animais , Linhagem Celular , Linhagem Celular Tumoral , Matriz Extracelular/patologia , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Regeneração Nervosa/fisiologia , Microambiente Tumoral/fisiologia , Cicatrização/fisiologia
5.
RSC Adv ; 8(71): 40639-40646, 2018 Dec 04.
Artigo em Inglês | MEDLINE | ID: mdl-35557887

RESUMO

Ship emulsified oil wastewater was used as the research object in this study. The highly efficient coagulant demulsification degreasing mechanism and microbubble flotation technology were combined and the effects of coagulant type and dosage amount on the demulsification of emulsified oil wastewater were evaluated. The influence of the mixed coagulation effect of pH values, temperature, and hydraulic condition parameters were determined and water intake, air intake, and oil content were regulated. The coagulant for the demulsification of emulsified oil wastewater was screened; the dosage was 500 mg L-1, and the removal capacity of the coagulant was in the following order: polyaluminum ferric chloride (PAFC) > polyaluminum chloride (PAC) > polysilicate aluminum ferric sulfate (PSAFS) > alum > Al2(SO4)3 > polyferric sulfate > FeCl3. Polyacrylamide (PAM) with added water was used to further reduce the oil content. The PAFC, PAC, and PSAFS were selected as coagulation-air flotation dynamic test alternative agents. The investment quantities of PAFC, PSAFS and PAM were 300 mg L-1, 300 mg L-1 and 30 mg L-1, respectively. The stirring time was 5 min, the pH value was 6.5-6.9, the flow rate was 0.25 m3 h-1, the oil content of the emulsified oil wastewater was 3000-5000 mg L-1 and the effluent oil was stable below 15 ppm. The microbubble generation device using air flotation effluent was used in the two air flotation treatments to enhance the device efficiency. The air flotation device adopted the structural design of the upper part of the water inlet and the lower part of the micro-air bubble, which can increase the collision probability of the microbubble and improve the efficiency of oil removal.

6.
Cancer Immunol Immunother ; 67(3): 353-366, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29116372

RESUMO

High-mobility group box 1 (HMGB1) is involved in the tumor-associated activation of regulatory T cells (Treg), but the mechanisms remain unknown. In a mouse tumor model, silencing HMGB1 in tumor cells or inhibiting tumor-derived HMGB1 not only dampened the capacity of tumor cells to produce thymic stromal lymphopoietin (TSLP), but also aborted the tumor-associated modulation of Treg-activating DC. Tumor-derived HMGB1 triggered the production of TSLP by tumor cells. Importantly, both tumor-derived HMGB1 and TSLP were necessary for modulating DC to activate Treg in a TSLP receptor (TSLPR)-dependent manner. In the therapeutic model, intratumorally inhibiting tumor-derived HMGB1 (causing downstream loss of TSLP production) attenuated Treg activation, unleashed tumor-specific CD8 T cell responses, and elicited CD8α+/CD103+DC- and T cell-dependent antitumor activity. These results suggest a new pathway for the activation of Treg involving in tumor-derived HMGB1 and TSLP, and have important implications for incorporating HMGB1 inhibitors into cancer immunotherapy.


Assuntos
Citocinas/fisiologia , Células Dendríticas/fisiologia , Proteína HMGB1/fisiologia , Ativação Linfocitária , Neoplasias Experimentais/imunologia , Linfócitos T Reguladores/imunologia , Animais , Feminino , Proteína HMGB1/antagonistas & inibidores , Imunoterapia , Camundongos , Neoplasias Experimentais/terapia , Linfopoietina do Estroma do Timo
7.
Oncoimmunology ; 6(6): e1315487, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28680744

RESUMO

The progressive tumor microenvironment (TME) coordinately supports tumor cell expansion and metastasis, while it antagonizes the survival and (poly-)functionality of antitumor T effector cells. There remains a clear need to develop novel therapeutic strategies that can transform the TME into a pro-inflammatory niche that recruits and sustains protective immune cell populations. While intravenous treatment with tumor-primed CD4+ T cells combined with intraperitoneal delivery of agonist anti-glucocorticoid-induced TNF receptor (α-GITR) mAb results in objective antitumor responses in murine early stage disease models, this approach is ineffective against more advanced tumors. Further subcutaneous co-administration of a vaccine consisting of tumor antigen-loaded dendritic cells (DC) failed to improve the antitumor efficacy of this approach. Remarkably, these same three therapeutic agents elicited significant antitumor benefits when the antitumor CD4+ T cells and tumor antigen-loaded DC were co-injected directly into tumors along with intratumoral or intraperitoneal delivery of α-GITR mAb. This latter protocol induced the production of an array of antitumor cytokines and chemokines within the TME, supporting increased tumor-infiltration by antitumor CD8+ T cells capable of mediating tumor regression and extended overall survival.

8.
J Immunol ; 194(12): 5937-47, 2015 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-25972487

RESUMO

The development of effective cancer vaccines remains an urgent, but as yet unmet, clinical need. This deficiency is in part due to an incomplete understanding of how to best invoke dendritic cells (DC) that are crucial for the induction of tumor-specific CD8(+) T cells capable of mediating durable protective immunity. In this regard, elevated expression of the transcription factor X box-binding protein 1 (XBP1) in DC appears to play a decisive role in promoting the ability of DC to cross-present Ags to CD8(+) T cells in the therapeutic setting. Delivery of DNA vaccines encoding XBP1 and tumor Ag to skin DC resulted in increased IFN-α production by plasmacytoid DC (pDC) from skin/tumor draining lymph nodes and the cross-priming of Ag-specific CD8(+) T cell responses associated with therapeutic benefit. Antitumor protection was dependent on cross-presenting Batf3(+) DC, pDC, and CD8(+) T cells. CD103(+) DC from the skin/tumor draining lymph nodes of the immunized mice appeared responsible for activation of Ag-specific naive CD8(+) T cells, but were dependent on pDC for optimal effectiveness. Similarly, human XBP1 improved the capacity of human blood- and skin-derived DC to activate human T cells. These data support an important intrinsic role for XBP1 in DC for effective cross-priming and orchestration of Batf3(+) DC-pDC interactions, thereby enabling effective vaccine induction of protective antitumor immunity.


Assuntos
Vacinas Anticâncer/imunologia , Apresentação Cruzada/imunologia , Células Dendríticas/imunologia , Neoplasias/imunologia , Vacinas de DNA/imunologia , Animais , Apresentação de Antígeno , Antígenos CD/metabolismo , Antígenos de Neoplasias/genética , Antígenos de Neoplasias/imunologia , Fatores de Transcrição de Zíper de Leucina Básica/metabolismo , Medula Óssea/imunologia , Medula Óssea/patologia , Vacinas Anticâncer/genética , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/imunologia , Células Dendríticas/metabolismo , Modelos Animais de Doenças , Feminino , Proteínas de Choque Térmico HSP72/genética , Proteínas de Choque Térmico HSP72/imunologia , Humanos , Cadeias alfa de Integrinas/metabolismo , Interferon-alfa/biossíntese , Oxirredutases Intramoleculares/genética , Oxirredutases Intramoleculares/imunologia , Neoplasias Pulmonares/imunologia , Neoplasias Pulmonares/secundário , Ativação Linfocitária , Melanoma Experimental , Camundongos , Camundongos Knockout , Neoplasias/mortalidade , Neoplasias/patologia , Neoplasias/terapia , Fenótipo , Ratos , Fatores de Transcrição de Fator Regulador X , Proteínas Repressoras/metabolismo , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/metabolismo , Receptor 3 Toll-Like/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/imunologia , Proteína 1 de Ligação a X-Box
9.
Oncoimmunology ; 3(10): e959321, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25941586

RESUMO

IL-15 supports improved antitumor immunity. How to best incorporate IL-15 into vaccine formulations for superior cancer immunotherapy remains a challenge. DC-derived IL-15 (DCIL-15) notably has the capacity to activate DC, to substitute for CD4+ Th and to potentiate vaccine efficacy making IL-15-based therapies attractive treatment options. We observed in transplantable melanoma, glioma and metastatic breast carcinoma models that DCIL-15-based DNA vaccines in which DC specifically express IL-15 and simultaneously produce tumor Aghsp70 were able to mediate potent therapeutic efficacy that required both host Batf3+ DC and CD8+ T cells. In an inducible BrafV600E/Pten-driven murine melanoma model, DCIL-15 (not rIL-15)-based DNA vaccines elicited durable therapeutic CD8+ T cell-dependent antitumor immunity. DCIL-15 was found to be superior to rIL-15 in "licensing" both mouse and human DC, and for activating CD8+ T cells. Such activation occurred even in the presence of Treg, without a need for CD4+ Th, but was IL-15/IL-15Rα-dependent. A single low-dose of DCIL-15 (not rIL-15)-based DC vaccines induced therapeutic antitumor immunity. CD14+ DC emigrating from human skin explants genetically-immunized by IL-15 and Aghsp70 were more effective than similar DC emigrating from the explants genetically-immunized by Aghsp70 in the presence of rIL-15 in expressing membrane-bound IL-15/IL-15Rα and activating CD8+ T cells. These results support future clinical use of DCIL-15 as a therapeutic agent in battling cancer.

10.
Mol Ther ; 20(2): 432-42, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21934655

RESUMO

In vivo dendritic cells (DC) targeting is an attractive approach with potential advantages in vaccine efficacy, cost, and availability. Identification of molecular adjuvants to in vivo "modulate " DC to coordinately render improved Th1 and CD8 T cell immunity, and attenuated deleterious Treg effects, is a critical challenge. Here, we report that in vivo genetic targeting of the active transcription factor XBP1s to DC (XBP1s/DC) potentiated vaccine-induced prophylactic and therapeutic antitumor immunity in multiple tumor models. This immunization strategy is based on a genetic vaccine encoding both cytomegalovirus (CMV)-driven vaccine Aghsp70 and DC-specific CD11c-driven XBP1s. The novel targeted vaccine induced durable Th1 and CD8 T cell responses to poorly immunogenic self/tumor antigen (Ag) and attenuated tumor-associated Treg suppressive function. Bone marrow (BM)-derived DC genetically modified to simultaneously overexpress XBP1s and express Aghsp70 upregulated CD40, CD70, CD86, interleukin (IL)-15, IL-15Rα, and CCR7 expression, and increased IL-6, IL-12, and tumor necrosis factor (TNF)-α production in vitro. XBP1s/DC elevated functional DEC205(+)CD8α(+)DC in the draining lymph nodes (DLN). The data suggest a novel role for XBP1s in modulating DC to potentiate tumor vaccine efficacy via overcoming two major obstacles to tumor vaccines (i.e., T cell hyporesponsiveness against poorly immunologic self/tumor Ag and tumor-associated Treg-mediated suppression) and improving DEC205(+)CD8α(+)DC.


Assuntos
Vacinas Anticâncer/genética , Vacinas Anticâncer/imunologia , Proteínas de Ligação a DNA/genética , Células Dendríticas/imunologia , Marcação de Genes , Neoplasias Experimentais/imunologia , Fatores de Transcrição/genética , Animais , Linfócitos T CD8-Positivos/imunologia , Vacinas Anticâncer/uso terapêutico , Linhagem Celular Tumoral , Sobrevivência Celular/genética , Proteínas de Ligação a DNA/metabolismo , Células Dendríticas/metabolismo , Feminino , Expressão Gênica , Ordem dos Genes , Interferon gama/biossíntese , Linfonodos/imunologia , Linfonodos/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Neoplasias Experimentais/tratamento farmacológico , Neoplasias Experimentais/mortalidade , Fatores de Transcrição de Fator Regulador X , Análise de Sobrevida , Linfócitos T Reguladores , Canais de Cátion TRPC/imunologia , Células Th1/imunologia , Fatores de Transcrição/metabolismo , Vacinas de DNA/genética , Vacinas de DNA/imunologia , Proteína 1 de Ligação a X-Box
11.
J Immunol ; 187(1): 118-25, 2011 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-21642542

RESUMO

Although high mobility group box 1 (HMGB1) in tumor cells is involved in many aspects of tumor progression, its role in tumor immune suppression remains elusive. Host cell-derived IL-10 suppressed a naturally acquired CD8 T cell-dependent antitumor response. The suppressive activity of tumor-associated Foxp3(+)CD4(+)CD25(+) regulatory T cells (Treg) was IL-10 dependent. Neutralizing HMGB1 impaired tumor cell-promoted IL-10 production by Treg. Short hairpin RNA-mediated knockdown of HMGB1 (HMGB1 KD) in tumor cells did not affect tumor cell growth but uncovered naturally acquired long-lasting tumor-specific IFN-γ- or TNF-α-producing CD8 T cell responses and attenuated their ability to induce Treg, leading to naturally acquired CD8 T cell- or IFN-γ-dependent tumor rejection. The data suggest that tumor cell-derived HMGB1 may suppress naturally acquired CD8 T cell-dependent antitumor immunity via enhancing Treg to produce IL-10, which is necessary for Treg-mediated immune suppression.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/patologia , Diferenciação Celular/imunologia , Proteína HMGB1/deficiência , Proteína HMGB1/genética , Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/patologia , Evasão Tumoral/imunologia , Animais , Antígenos de Neoplasias/biossíntese , Antígenos de Neoplasias/genética , Antígenos de Neoplasias/fisiologia , Linfócitos T CD8-Positivos/metabolismo , Diferenciação Celular/genética , Linhagem Celular Tumoral , Neoplasias do Colo/imunologia , Neoplasias do Colo/patologia , Neoplasias do Colo/prevenção & controle , Feminino , Técnicas de Silenciamento de Genes , Proteína HMGB1/fisiologia , Humanos , Imunidade Inata/genética , Interleucina-10/biossíntese , Interleucina-10/genética , Interleucina-10/fisiologia , Neoplasias Mamárias Experimentais/imunologia , Neoplasias Mamárias Experimentais/patologia , Neoplasias Mamárias Experimentais/prevenção & controle , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/farmacologia , Linfócitos T Reguladores/metabolismo , Evasão Tumoral/genética
12.
Mol Ther ; 17(7): 1274-81, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19417739

RESUMO

Tumor-primed CD4(+) T cells from splenocytes of tumor-rejection mice in combination with in vivo glucocorticoid-induced tumor necrosis factor receptor (GITR) ligation (the combination therapy) elicited effective host CD8(+) T cell-dependent therapeutic immunity against a murine breast tumor. GITR ligation in vitro enhanced tumor-primed CD4(+) T-cell activity and partially abrogated regulatory T cells (Treg) suppressor function. Dendritic cells (DCs) from tumor-draining lymph nodes (TDLNs) of tumor-bearing mice treated by the combination therapy stimulated Ag-specific T cells and produced interleukin (IL)-12 ex vivo. Whereas tumor-primed CD4(+) T cells or in vivo GITR ligation alone induced a tumor-specific interferon (IFN)-gamma-producing cellular response, the combination therapy enhanced and sustained it. Furthermore, the combination therapy in vivo attenuated Treg's ability to suppress IL-12 production by DCs and IFN-gamma production by effectors ex vivo. Importantly, tumor-primed CD4(+) CD25(-) T cells from splenocytes of untreated tumor-bearing mice in combination with in vivo GITR ligation also elicited an effective therapeutic effect in this model. These data suggest that the combination therapy may improve DC function, accentuate tumor-specific T-cell responses, and attenuate Treg suppressor function, thereby eliciting effective therapeutic immunity.


Assuntos
Neoplasias da Mama/imunologia , Neoplasias da Mama/terapia , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/transplante , Proteína Relacionada a TNFR Induzida por Glucocorticoide/metabolismo , Imunoterapia Adotiva/métodos , Animais , Anticorpos Monoclonais/uso terapêutico , Linfócitos T CD8-Positivos/imunologia , Células Dendríticas/imunologia , Feminino , Imunidade Celular/efeitos dos fármacos , Imunidade Celular/imunologia , Interferon gama/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Receptores do Fator de Necrose Tumoral/imunologia , Linfócitos T Reguladores/imunologia
13.
J Immunol ; 182(10): 6160-7, 2009 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-19414769

RESUMO

Regulatory T cell (Treg) from mice bearing a breast tumor were elevated (tumor Treg). In vitro, whereas tumor Treg ability to inhibit tumor-primed CD4(+) T cell activity is comparable to Treg from naive mice (naive Treg), only tumor Treg suppress naive CD8(+) T cell activation and DC function. Neither tumor Treg nor naive Treg can suppress antitumor immunity at the effector phase of the immune response induced by adoptively transferred tumor-primed CD4(+) T cells. This is consistent with the observation that, in this model, neither tumor Treg nor naive Treg can inhibit effectors in vitro or in vivo. However, tumor Treg abrogate tumor-specific CD8(+) T cell responses in tumor-draining lymph nodes and antitumor immunity at the early stage of the immune response induced by adoptively transferred tumor-primed CD4(+) T cells. These data indicate that, in this model, tumor Treg potently abrogate tumor-specific CD8(+) T cell responses in tumor-draining lymph nodes, thereby suppressing antitumor immunity at the early stage of the immune response induced by adoptively transferred tumor-primed CD4(+) T cells.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Neoplasias Mamárias Experimentais/imunologia , Linfócitos T Reguladores/imunologia , Transferência Adotiva , Animais , Feminino , Ativação Linfocitária , Camundongos , Camundongos Endogâmicos BALB C
14.
J Immunol ; 181(6): 4363-70, 2008 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-18768895

RESUMO

Administration of anti-CD25 mAb before an aggressive murine breast tumor inoculation provoked effective antitumor immunity. Compared with CD4(+) T cells purified from anti-CD25 mAb-pretreated mice that did not reject tumor, CD4(+) T cells purified from anti-CD25 mAb-pretreated mice that rejected tumor stimulated by dendritic cells (DCs) produced more IFN-gamma and IL-2, and less IL-17 in vitro, and ignited protective antitumor immunity in vivo in an adoptive transfer model. Tumor Ag-loaded DCs activated naive CD8(+) T cells in the presence of these CD4(+) T cells in vitro. Tumor Ag and adoptively transferred CD4(+) T cells were both required for inducing a long-term tumor-specific IFN-gamma-producing cellular response and potent protective antitumor activity. Although adoptively transferred CD4(+) T cells ignited effective tumor-specific antitumor immunity in wild-type mice, they failed to do so in endogenous NK cell-depleted, Gr-1(+) cell-depleted, CD40(-/-), CD11c(+) DC-depleted, B cell(-/-), CD8(+) T cell-depleted, or IFN-gamma(-/-) mice. Collectively, the data suggest that adoptively transferred CD4(+) T cells orchestrate both endogenous innate and adaptive immunity to generate effective tumor-specific long-term protective antitumor immunity. The data also demonstrate the pivotal role of endogenous DCs in the tumor-specific protection ignited by adoptively transferred CD4(+) T cells. Thus, these findings highlight the importance of adoptively transferred CD4(+) T cells, as well as host immune components, in generating effective tumor-specific long-term antitumor activity.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/transplante , Imunoterapia Adotiva/métodos , Neoplasias Mamárias Animais/imunologia , Neoplasias Mamárias Animais/terapia , Animais , Anticorpos Monoclonais/administração & dosagem , Anticorpos Monoclonais/uso terapêutico , Linfócitos T CD4-Positivos/metabolismo , Linhagem Celular Tumoral , Relação Dose-Resposta Imunológica , Feminino , Rejeição de Enxerto/imunologia , Interferon gama/biossíntese , Subunidade alfa de Receptor de Interleucina-2/imunologia , Neoplasias Mamárias Animais/patologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Camundongos Transgênicos , Invasividade Neoplásica , Transplante de Neoplasias
15.
J Immunol ; 179(3): 1960-8, 2007 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-17641063

RESUMO

Metastatic disease is the major cause of morbidity and mortality in cancer. Although surgery, chemotherapy, or radiation can often control primary tumor growth, successful eradication of disseminated metastases remains rare. We have now tested whether direct targeting tumor tissues to generate antitumor immune response before surgical excision produces sufficient CTL against micrometastases. One unsolved problem is whether such response allows coming CTL to be educated and then exit the tumor site. Another unsolved problem is whether these CTL can then patrol and effectively eliminate spontaneously metastasized tumor cells in the periphery. In this study, we have shown that adenovirus-expressing TNFSF14 [LIGHT (name derived from homologous to lymphotoxins, shows inducible expression, and competes with herpes simplex virus glycoprotein D for herpes virus entry mediator, a receptor expressed by T lymphocytes); Ad-LIGHT] inoculated directly into primary 4T1 tumor, a highly aggressive, spontaneously metastasizing mammary carcinoma, followed by surgical removal of the primary tumor can eradicate established and disseminated metastatic tumor cells in the peripheral tissues. Furthermore, we clearly show with a fibrosarcoma model Ag104L(d) that local treatment can generate plenty of tumor-specific CTL that exit the primary tumor and infiltrate distal tumors to completely eradicate distal tumors. Therefore, targeting the primary tumor with Ad-LIGHT before surgical excision is a new strategy to elicit better immune response for the eradication of spontaneous metastases.


Assuntos
Citotoxicidade Imunológica , Neoplasias Pulmonares/imunologia , Neoplasias Pulmonares/secundário , Neoplasias Mamárias Experimentais/imunologia , Neoplasias Mamárias Experimentais/terapia , Linfócitos T Citotóxicos/imunologia , Adenoviridae/genética , Animais , Antígenos de Neoplasias/imunologia , Linhagem Celular Tumoral , Neoplasias do Colo/imunologia , Neoplasias do Colo/patologia , Neoplasias do Colo/prevenção & controle , Citotoxicidade Imunológica/genética , Feminino , Vetores Genéticos/fisiologia , Rejeição de Enxerto/genética , Rejeição de Enxerto/imunologia , Humanos , Imunidade Inata/genética , Neoplasias Pulmonares/prevenção & controle , Ativação Linfocitária/genética , Ativação Linfocitária/imunologia , Neoplasias Mamárias Experimentais/patologia , Melanoma Experimental/imunologia , Melanoma Experimental/patologia , Melanoma Experimental/prevenção & controle , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Linfócitos T/imunologia , Linfócitos T/patologia , Linfócitos T Citotóxicos/citologia , Membro 14 da Superfamília de Ligantes de Fatores de Necrose Tumoral/genética , Membro 14 da Superfamília de Ligantes de Fatores de Necrose Tumoral/fisiologia
16.
Hum Gene Ther ; 16(11): 1255-66, 2005 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-16259559

RESUMO

Direct in vivo administration of a lentiviral vaccine has been shown to transduce dendritic cells (DCs) in order to induce antigen-specific CD8+ T cell responses, but the efficacy of antitumor immunity has not been reported. In this study we tested whether direct in vivo administration of a lentiviral vaccine can induce selfantigen- based therapeutic antitumor immunity in murine tumor models. Lentiviral vector (LV) transduced DCs efficiently in vitro and was able to transduce DCs in vivo. LV-transduced DCs effectively presented antigens to T cells. Compared with a naked DNA tyrosinase-related protein-2 (TRP2)-heat shock protein-70 (hsp70) vaccine, the TRP2-specific interferon-gamma-producing CD8+ T cell response was augmented by direct in vivo administration of an LV-TRP2hsp70 vaccine, which induced significant therapeutic antitumor immunity in subcutaneous B16 and subcutaneous GL-26 models. Moreover, in vivo administration of an LV-NeuEDhsp70 vaccine induced significant therapeutic antitumor immunity against spontaneous breast tumors in a BALB/c- Neu transgenic model. Our observations indicate that direct in vivo administration of a lentiviral vaccine not only enhances antigen-specific CD8+ T cell responses, but also generates significant therapeutic antitumor activities.


Assuntos
Lentivirus/genética , Melanoma Experimental/imunologia , Vacinas Virais/imunologia , Animais , Sequência de Bases , Linfócitos T CD8-Positivos/imunologia , Linhagem Celular Tumoral , Primers do DNA , Células Dendríticas/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Vacinas Virais/administração & dosagem
17.
Mol Ther ; 11(6): 941-9, 2005 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15922965

RESUMO

The rapid growth and ruthless metastasis of tumors demand effective broad immune responses. Dendritic cells (DCs) are critical in developing tumor vaccines. Recent investigations have been focused on modifying tumor antigens to target DCs to induce immune responses efficiently in vivo. In this study, human hsp70 was fused to the extracellular domain of rat Her2/Neu (NeuEDhsp70) for enhancing anti-tumor immunity in aggressive breast tumor models. NeuEDhsp70-conditioned DCs produced significant IL-12p40 and effectively presented NeuED antigens to T cells in vitro. NeuEDhsp70 DNA vaccine induced enhanced Neu-specific antibody and IFN-gamma-producing cellular immune responses in vivo. Although NeuEDhsp70 and NeuED DNA vaccines elicited comparable therapeutic anti-tumor immunity in an aggressive primary breast tumor model, NeuEDhsp70 DNA vaccine significantly increased survival and reduced metastasis in an aggressive spontaneous metastatic breast tumor model. Results from animal experiments with depletion of immune cells or with deficiency of CD40 molecules indicate that T cells and CD40 molecules are critical in the anti-tumor immunity induced by NeuEDhsp70 DNA vaccine. These observations suggest that NeuEDhsp70 DNA vaccine is a promising reagent to induce potent anti-tumor immunity to an aggressive spontaneous metastatic breast tumor.


Assuntos
Neoplasias da Medula Óssea/prevenção & controle , Vacinas Anticâncer/imunologia , Vacinas Anticâncer/uso terapêutico , Neoplasias Mamárias Animais/tratamento farmacológico , Proteínas Recombinantes de Fusão/imunologia , Animais , Antígenos de Neoplasias/imunologia , Neoplasias da Medula Óssea/secundário , Linfócitos T CD4-Positivos/imunologia , Antígenos CD40/análise , Vacinas Anticâncer/genética , Células Dendríticas/imunologia , Feminino , Interleucina-12/análise , Subunidade p40 da Interleucina-12 , Neoplasias Mamárias Animais/imunologia , Neoplasias Mamárias Animais/patologia , Camundongos , Subunidades Proteicas/análise , Ratos , Receptor ErbB-2/imunologia , Proteínas Recombinantes de Fusão/genética , Vacinas de DNA/imunologia , Vacinas de DNA/uso terapêutico
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA