Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 285
Filtrar
1.
Int J Pharm ; 638: 122932, 2023 May 10.
Artigo em Inglês | MEDLINE | ID: mdl-37031810

RESUMO

Heterologous prime-boost vaccines have the potential to promote higher immune responses than homologous prime-boost vaccines and were used in this murine study to investigate the effect on the magnitude of the cellular (and humoral) antigen-specific immune responses and antitumor efficacy when a microparticle formulation (prime) is combined with an adenoviral vaccine (boost). Specifically, the prime comprised chick egg ovalbumin (OVA; 25 µg/dose), used here as a model tumor antigen (TA), encapsulated in microparticles (∼700 nm diameter) made from the biodegradable polymer, 50:50 poly(lactic-co-glycolic acid) (PLGA); while attenuated adenovirus (type 5) encoding OVA (Ad5OVA; 108 PFU/dose) was employed as the boost. The ability of OVA-loaded microparticles to enhance OVA-specific antibody responses, OVA-specific CD3 + CD8 + T cell responses and antitumor activity (i.e., protection against OVA-expressing tumor-challenge) to the heterologous prime-boost vaccine was investigated; and it was found that this prime-boost combination could significantly enhance OVA-specific cellular responses compared to all other vaccination groups and was the only group to confer a significant survival advantage over the unvaccinated group (naïve) in a prophylactic animal tumor model. This finding illustrates the potential for combining TA-loaded PLGA-based microparticles with other vaccine formats to improve tumor-specific cellular immune responses.


Assuntos
Neoplasias , Vacinas Virais , Camundongos , Animais , Adenoviridae/genética , Antígenos , Imunidade Celular
2.
Acta Biomater ; 158: 525-534, 2023 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-36572250

RESUMO

Codelivering subunit antigens and Toll-like receptor (TLR) molecular adjuvants via nanocarriers can stimulate potent innate and specific immune responses. Simple and effective nanovaccines fabrication is crucial for application. However, most nanovaccines were fabricated by introducing additional delivery materials, increasing safety risk, cost and processing complexity. Herein, a carrier-free nanovaccine was facilely prepared using a TLR1/TLR2 adjuvant, Diprovocim, rich in benzene rings that could interact with aromatic residues in subunit antigens through π-π stacking without additional materials. The carrier-free nanovaccines with a narrow size distribution could target lymph nodes (LNs) after intravenous injection to mice. The carrier-free nanovaccines based on ovalbumin (OVA) can stimulate strong antibody titers and CD4+ and CD8+ T cell immune responses in mice, and it synergized with anti-PD1 showing a potent tumor suppression in B16F10-OVA tumor model of mice. Furthermore, the carrier-free nanovaccine with glycoprotein E (gE), a glycoprotein of the varicella-zoster virus (VZV), also showed potent humoral and cellular immune responses. Therefore, using subunit proteins to support Diprovocim by π-π stacking provides a new approach for the preparation and application of novel vaccines for tumor therapy and prevention of infectious diseases. STATEMENT OF SIGNIFICANCE: Codelivering subunit antigens and adjuvants via nanocarriers stimulate potent innate and specific immune responses. However, existing delivery materials for fabricating nanovaccines will inevitably increase the cost of preparation, controllability, process complexity and safety assessment. Therefore, this study easily prepared carrier-free nanovaccines using the benzene ring-rich TLR1/TLR2 adjuvant Diprovocim, which can interact with aromatic residues in subunit antigens via π-π stacking without additional materials. The carrier-free nanovaccines of OVA demonstrated a potent tumor inhibition in treating melanoma in combination with anti-PD1. And the nanovaccines of gE stimulated a strong antibody titer and cellular immune response for herpes zoster. Thus, the present study provides a new approach for the preparation of subunit vaccines to combat various cancers and virus infections.


Assuntos
Nanopartículas , Neoplasias , Viroses , Animais , Camundongos , Receptor 1 Toll-Like , Receptor 2 Toll-Like , Benzeno , Imunidade Celular , Adjuvantes Imunológicos/química , Antígenos , Vacinas de Subunidades , Nanopartículas/química
3.
Front Immunol ; 12: 715234, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34354714

RESUMO

A paradigm shift in the understanding of the exhausted CD8+ T cell (Tex) lineage is underway. Originally thought to be a uniform population that progressively loses effector function in response to persistent antigen, single-cell analysis has now revealed that CD8+ Tex is composed of multiple interconnected subpopulations. The heterogeneity within the CD8+ Tex lineage is comprised of immune checkpoint blockade (ICB) permissive and refractory subsets termed stem-like and terminally differentiated cells, respectively. These populations occupy distinct peripheral and intratumoral niches and are characterized by transcriptional processes that govern transitions between cell states. This review presents key findings in the field to construct an updated view of the spatial, transcriptional, and functional heterogeneity of anti-tumoral CD8+ Tex. These emerging insights broadly call for (re-)focusing cancer immunotherapies to center on the driver mechanism(s) underlying the CD8+ Tex developmental continuum aimed at stabilizing functional subsets.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Imunidade , Neoplasias/imunologia , Animais , Antígenos/imunologia , Biomarcadores , Linfócitos T CD8-Positivos/efeitos dos fármacos , Linfócitos T CD8-Positivos/metabolismo , Gerenciamento Clínico , Suscetibilidade a Doenças , Epigênese Genética , Interações Hospedeiro-Patógeno/imunologia , Humanos , Inibidores de Checkpoint Imunológico/farmacologia , Proteínas de Checkpoint Imunológico/genética , Proteínas de Checkpoint Imunológico/metabolismo , Terapia de Alvo Molecular , Neoplasias/metabolismo , Neoplasias/patologia , Neoplasias/terapia , Especificidade de Órgãos , Análise de Célula Única , Transcrição Gênica
4.
Front Immunol ; 12: 688753, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34276678

RESUMO

Objectives: Type I interferons (IFNs) are central and reflective of disease activity in systemic lupus erythematosus (SLE). However, IFN-α levels are notoriously difficult to measure and the type I IFN gene signature (IGS) is not yet available in clinical routine. This study evaluates galectin-9 and an array of chemokines/cytokines in their potential as surrogate markers of type I IFN and/or SLE disease activity. Methods: Healthy controls and well-characterized Swedish SLE patients from two cross-sectional cohorts (n=181; n=59) were included, and a subgroup (n=21) was longitudinally followed. Chemokine/cytokine responses in immune complex triggered IFN-α activity was studied in healthy donor peripheral blood mononuclear cells (PBMC). Levels of chemokines/cytokines and galectin-9 were measured by immunoassays. Gene expression was quantified by qPCR. Results: The IGS was significantly (p<0.01) correlated with galectin-9 (rho=0.54) and CXCL10 (rho=0.37) levels whereas serum IFN-α correlated with galectin-9 (rho=0.36), CXCL10 (rho=0.39), CCL19 (rho=0.26) and CCL2 (rho=0.19). The strongest correlation was observed between galectin-9 and TNF (rho=0.56). IFN-α and disease activity (SLEDAI-2K) were correlated (rho=0.20) at cross-sectional analysis, but no significant associations were found between SLEDAI-2K and galectin-9 or chemokines. Several inflammatory mediators increased at disease exacerbation although CCL19, CXCL11, CXCL10, IL-10 and IL-1 receptor antagonist were most pronounced. Immune complex-stimulation of PBMC increased the production of CCL2, CXCL8 and TNF. Conclusion: Galectin-9 and CXCL10 were associated with type I IFN in SLE but correlated stronger with TNF. None of the investigated biomarkers showed a convincing association with disease activity, although CXCL10 and CCL19 performed best in this regard.


Assuntos
Interferon-alfa/sangue , Leucócitos Mononucleares/metabolismo , Lúpus Eritematoso Sistêmico/diagnóstico , Transcriptoma , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Antígenos/genética , Biomarcadores/sangue , Estudos de Casos e Controles , Células Cultivadas , Quimiocina CCL19/metabolismo , Quimiocina CXCL10/metabolismo , Quimiocinas/metabolismo , Estudos Transversais , Proteínas do Citoesqueleto/genética , Feminino , Galectinas/metabolismo , Perfilação da Expressão Gênica , Humanos , Imunoensaio , Leucócitos Mononucleares/imunologia , Estudos Longitudinais , Lúpus Eritematoso Sistêmico/sangue , Lúpus Eritematoso Sistêmico/genética , Lúpus Eritematoso Sistêmico/imunologia , Masculino , Proteínas de Membrana/genética , Pessoa de Meia-Idade , Oxirredutases atuantes sobre Doadores de Grupo CH-CH , Valor Preditivo dos Testes , Proteínas/genética , Suécia , Proteínas Supressoras de Tumor/genética , Adulto Jovem
5.
Mol Cell Proteomics ; 20: 100022, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33583769

RESUMO

The approach of peptide-based anticancer vaccination has proven the ability to induce cancer-specific immune responses in multiple studies for various cancer entities. However, clinical responses remain so far limited to single patients and broad clinical applicability was not achieved. Therefore, further efforts are required to improve peptide vaccination in order to integrate this low-side-effect therapy into the clinical routine of cancer therapy. To design clinically effective peptide vaccines in the future, different issues have to be addressed and optimized comprising antigen target selection as well as choice of optimal adjuvants and vaccination schedules. Furthermore, the combination of peptide-based vaccines with other immuno- and molecular targeted therapies as well as the development of predictive biomarkers could further improve efficacy. In this review, current approaches in the development of peptide-based vaccines and critical implications for optimal vaccine design are discussed.


Assuntos
Vacinas Anticâncer/administração & dosagem , Neoplasias/terapia , Vacinas de Subunidades/administração & dosagem , Adjuvantes Imunológicos/administração & dosagem , Antígenos/imunologia , Biomarcadores , Humanos , Neoplasias/imunologia
6.
Int J Mol Sci ; 22(3)2021 Jan 22.
Artigo em Inglês | MEDLINE | ID: mdl-33499003

RESUMO

Radiotherapy remains one of the contemporary cornerstones of cancer treatment in the neoadjuvant, curative, adjuvant and palliative settings, either in isolation or as a multimodal approach. Moreover, recent advances in targeted immune checkpoint therapy have firmly established immunotherapy as the fourth pillar in cancer therapy alongside surgery, chemotherapy and notably radiotherapy. There is emerging evidence to suggest both radioresistance and reduced efficacy of immune checkpoint blockade (ICB) are potentiated by the tumour microenvironment (TME) and in fact modulating aspects of this immunosuppressive milieu is instrumental to unlocking anti-tumour immunity. The response rates of Upper Gastrointestinal (UGI) malignancies to ICB remains modest at 10-15%, compared to melanoma at 20-40%. Harnessing the effects of radiotherapy through remodelling of the TME using ICB as a radiosensitisor is an avenue showing promise. Here we explore the rationale behind combining radiotherapy with ICB, as a symbiotic relationship in shifting the balance in favour of anti-tumour immunity. We discuss the effects of radiotherapy on immunogenic cell death, the concept of the abscopal effect, the importance of the cGAS STING pathway, and their relevance in the context of the tumour microenvironment. Furthermore, dosing and timing of radiotherapy and ICB is now being evaluated for its synergistic effects on host tumour immunity, and we review the ongoing efforts and current available literature for single agent and dual agent ICB in combination multimodal therapy for both locally advanced operable and metastatic disease of the upper gastrointestinal tract.


Assuntos
Neoplasias Gastrointestinais/radioterapia , Neoplasias Gastrointestinais/terapia , Imunoterapia/tendências , Animais , Antígenos/química , Biomarcadores , Morte Celular , Quimiorradioterapia , Terapia Combinada , Modelos Animais de Doenças , Fracionamento da Dose de Radiação , Neoplasias Gastrointestinais/imunologia , Humanos , Fatores Imunológicos/farmacologia , Inflamação , Melanoma/patologia , Proteínas de Membrana/metabolismo , Camundongos , Metástase Neoplásica , Nucleotidiltransferases/química , Neoplasias Cutâneas/patologia , Microambiente Tumoral/imunologia , Trato Gastrointestinal Superior/patologia , Trato Gastrointestinal Superior/efeitos da radiação
7.
HNO ; 68(2): 100-105, 2020 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-32006045

RESUMO

This manuscript describes the functional properties of the exosomes released from melanoma cells. It details the characteristics of the tumor antigen chondroitin sulfate proteoglycan 4 (CSPG4), which is used as a marker to separate exosomes released by melanoma cells from exosomes released by nonmalignant cells. The results are discussed in view of the potential role of melanoma cell-derived exosomes in the escape of malignant cells from the host's immune system.


Assuntos
Proteoglicanas de Sulfatos de Condroitina , Exossomos , Melanoma , Proteínas de Membrana , Neoplasias Cutâneas , Antígenos , Biomarcadores/análise , Líquidos Corporais , Proteoglicanas de Sulfatos de Condroitina/análise , Humanos , Melanoma/tratamento farmacológico , Melanoma/imunologia , Proteínas de Membrana/análise , Proteoglicanas , Neoplasias Cutâneas/tratamento farmacológico , Neoplasias Cutâneas/imunologia
8.
Sci Adv ; 5(10): eaaw6870, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31681841

RESUMO

Erythrocytes or red blood cells (RBCs) represent a promising cell-mediated drug delivery platform due to their inherent biocompatibility. Here, we developed an antigen delivery system based on the nanoerythrosomes derived from RBCs, inspired by the splenic antigen-presenting cell targeting capacity of senescent RBCs. Tumor antigens were loaded onto the nanoerythrosomes by fusing tumor cell membrane-associated antigens with nanoerythrosomes. This tumor antigen-loaded nanoerythrosomes (nano-Ag@erythrosome) elicited antigen responses in vivo and, in combination with the anti-programmed death ligand 1 (PD-L1) blockade, inhibited the tumor growth in B16F10 and 4T1 tumor models. We also generated a tumor model showing that "personalized nano-Ag@erythrosomes" could be achieved by fusing RBCs and surgically removed tumors, which effectively reduced tumor recurrence and metastasis after surgery.


Assuntos
Antígenos/imunologia , Eritrócitos/metabolismo , Imunoterapia , Nanopartículas/química , Neoplasias/imunologia , Neoplasias/terapia , Proteolipídeos/química , Animais , Células Apresentadoras de Antígenos/metabolismo , Antígeno B7-H1/metabolismo , Biomarcadores/metabolismo , Células Dendríticas/metabolismo , Macrófagos/metabolismo , Melanoma Experimental/imunologia , Melanoma Experimental/patologia , Melanoma Experimental/cirurgia , Melanoma Experimental/terapia , Camundongos Endogâmicos C57BL , Metástase Neoplásica , Recidiva Local de Neoplasia/patologia , Baço/patologia , Resultado do Tratamento
10.
Annu Rev Chem Biomol Eng ; 10: 337-359, 2019 06 07.
Artigo em Inglês | MEDLINE | ID: mdl-31173518

RESUMO

Although vaccines have been the primary defense against widespread infectious disease for decades, there is a critical need for improvement to combat complex and variable diseases. More control and specificity over the immune response can be achieved by using only subunit components in vaccines. However, these often lack sufficient immunogenicity to fully protect, and conjugation or carrier materials are required. A variety of protein and peptide biomaterials have improved effectiveness and delivery of subunit vaccines for infectious, cancer, and autoimmune diseases. They are biodegradable and have control over both material structure and immune function. Many of these materials are built from naturally occurring self-assembling proteins, which have been engineered for incorporation of vaccine components. In contrast, others are de novo designs of structures with immune function. In this review, protein biomaterial design, engineering, and immune functionality as vaccines or immunotherapies are discussed.


Assuntos
Materiais Biocompatíveis/química , Imunoterapia , Peptídeos/imunologia , Vacinas de Subunidades/imunologia , Antígenos/imunologia , Antígenos/metabolismo , Doenças Transmissíveis/imunologia , Doenças Transmissíveis/terapia , Humanos , Nanoestruturas/química , Neoplasias/imunologia , Neoplasias/terapia , Peptídeos/química , Peptídeos/metabolismo , Vacinas de Subunidades/química , Vacinas de Subunidades/metabolismo , Vacinas de Partículas Semelhantes a Vírus/imunologia
11.
Cancer Immunol Res ; 6(7): 870-880, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29792298

RESUMO

High-risk human papillomavirus-associated cancers express viral oncoproteins (e.g., E6 and E7) that induce and maintain the malignant phenotype. The viral origin of these proteins makes them attractive targets for development of a therapeutic vaccine. Camelid-derived single-domain antibody fragments (nanobodies or VHHs) that recognize cell surface proteins on antigen-presenting cells (APC) can serve as targeted delivery vehicles for antigens attached to them. Such VHHs were shown to induce CD4+ and CD8+ T-cell responses against model antigens conjugated to them via sortase, but antitumor responses had not yet been investigated. Here, we tested the ability of an anti-CD11b VHH (VHHCD11b) to target APCs and serve as the basis for a therapeutic vaccine to induce CD8+ T-cell responses against HPV+ tumors. Mice immunized with VHHCD11b conjugated to an H-2Db-restricted immunodominant E7 epitope (E749-57) had more E7-specific CD8+ T cells compared with those immunized with E749-57 peptide alone. These CD8+ T cells acted prophylactically and conferred protection against a subsequent challenge with HPV E7-expressing tumor cells. In a therapeutic setting, VHHCD11b-E749-57 vaccination resulted in greater numbers of CD8+ tumor-infiltrating lymphocytes compared with mice receiving E749-57 peptide alone in HPV+ tumor-bearing mice, as measured by in vivo noninvasive VHH-based immune-positron emission tomography (immunoPET), which correlated with tumor regression and survival outcome. Together, these results demonstrate that VHHs can serve as a therapeutic cancer vaccine platform for HPV-induced cancers. Cancer Immunol Res; 6(7); 870-80. ©2018 AACR.


Assuntos
Antígenos/imunologia , Neoplasias/etiologia , Papillomaviridae/imunologia , Infecções por Papillomavirus/complicações , Infecções por Papillomavirus/imunologia , Anticorpos de Domínio Único/imunologia , Animais , Biomarcadores , Vacinas Anticâncer/imunologia , Modelos Animais de Doenças , Feminino , Imunização , Imunoterapia , Camundongos , Camundongos Knockout , Modelos Biológicos , Neoplasias/diagnóstico , Neoplasias/metabolismo , Neoplasias/terapia , Proteínas Oncogênicas Virais/genética , Proteínas Oncogênicas Virais/imunologia , Infecções por Papillomavirus/virologia , Tomografia por Emissão de Pósitrons , Ligação Proteica
13.
Mol Ther ; 25(2): 504-511, 2017 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-28153097

RESUMO

Recombinant lentiviral vectors (LVs) are highly effective vaccination vehicles that elicit protective T cell immunity in disease models. Dendritic cells (DCs) acquire antigen at sites of vaccination and migrate to draining lymph nodes, where they prime vaccine-specific T cells. The potency with which LVs activate CD8+ T cell immunity has been attributed to the transduction of DCs at the immunization site and durable presentation of LV-encoded antigens. However, it is not known how LV-encoded antigens continue to be presented to T cells once directly transduced DCs have turned over. Here, we report that LV-encoded antigen is efficiently cross-presented by DCs in vitro. We have further exploited the temporal depletion of DCs in the murine CD11c.DTR (diphtheria toxin receptor) model to demonstrate that repopulating DCs that were absent at the time of immunization cross-present LV-encoded antigen to T cells in vivo. Indirect presentation of antigen from transduced cells by DCs is sufficient to prime functional effector T cells that control tumor growth. These data suggest that DCs cross-present immunogenic antigen from LV-transduced cells, thereby facilitating prolonged activation of T cells in the absence of circulating LV particles. These are findings that may impact on the future design of LV vaccination strategies.


Assuntos
Apresentação de Antígeno , Antígenos/imunologia , Apresentação Cruzada/imunologia , Células Dendríticas/imunologia , Imunidade Celular , Linfócitos T/imunologia , Linfócitos T/metabolismo , Animais , Antígenos/genética , Biomarcadores , Antígeno CD11c/metabolismo , Vacinas Anticâncer/imunologia , Técnicas de Cocultura , Células Dendríticas/metabolismo , Vetores Genéticos/genética , Fator de Crescimento Semelhante a EGF de Ligação à Heparina/imunologia , Lentivirus/genética , Linfonodos/imunologia , Linfonodos/metabolismo , Camundongos , Modelos Animais , Neoplasias/imunologia , Neoplasias/patologia , Neoplasias/terapia , Transdução Genética , Vacinas/genética , Vacinas/imunologia
14.
Eur J Immunol ; 46(7): 1574-82, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-27271222

RESUMO

T-cell metabolism is central to the shaping of a successful immune response. However, there are pathological situations where T cells are rendered dysfunctional and incapable of eliminating infected or transformed cells. Here, we review the current knowledge on T-cell metabolism and how persistent antigenic stimulation, in the form of cancer and chronic viral infection, modifies both metabolic and functional pathways in T cells.


Assuntos
Reprogramação Celular/imunologia , Metabolismo Energético/imunologia , Ativação Linfocitária/imunologia , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/metabolismo , Adaptação Biológica/genética , Adaptação Biológica/imunologia , Animais , Antígenos/imunologia , Reprogramação Celular/genética , Metabolismo Energético/genética , Regulação da Expressão Gênica , Interações Hospedeiro-Patógeno/genética , Interações Hospedeiro-Patógeno/imunologia , Humanos , Memória Imunológica , Ativação Linfocitária/genética , Neoplasias/imunologia , Neoplasias/metabolismo , Transdução de Sinais , Subpopulações de Linfócitos T/citologia , Viroses/imunologia , Viroses/metabolismo
15.
Nanomedicine ; 11(2): 293-300, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24780311

RESUMO

We have developed a quantitative approach to eventually enable precise and multiplexing protein analysis of very small systems, down to a single or a few cells. Through DNA-directed immobilization of DNA-protein conjugates we immobilized antibodies specific for a certain protein of interest, on a complementary DNA nanoarray fabricated by means of nanografting, a nanolithography technique based on atomic force microscopy (AFM). The proof of concept was realized for glial fibrillary acidic protein (GFAP), a biomarker crucial in cell's differentiation of astrocytes, and functional to grade classification of gliomas, the most common of primary malignant brain tumors. The efficiency of the nano-immuno sensing was tested by obtaining the immobilization of purified recombinant GFAP protein at different concentration in a standard solution then in a cellular lysate. A comparison of sensitivity between our technique and conventional ELISA assays is provided at the end of the paper. FROM THE CLINICAL EDITOR: This team developed a quantitative approach to enable precise and multiplexing protein analysis of very small systems, down to a single or a few cells, demonstrating the utility of this DNA-based nano-immunoassay in the detection of GFAP.


Assuntos
DNA/química , Proteína Glial Fibrilar Ácida/isolamento & purificação , Glioma/imunologia , Imunoensaio , Anticorpos/química , Anticorpos/imunologia , Antígenos/química , Antígenos/imunologia , Astrócitos/imunologia , Astrócitos/patologia , Biomarcadores/química , Proteína Glial Fibrilar Ácida/imunologia , Glioma/diagnóstico , Humanos , Microscopia de Força Atômica
16.
Talanta ; 132: 162-74, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25476294

RESUMO

Cancer and cardiovascular diseases are the major threats to global health. Hence, there is a growing demand for a range of portable, rapid and low cost biosensing devices for the detection of these diseases. Electrochemical immunosensors are simple, rapid, reliable and inexpensive devices and they have sensitive detection limits to monitor both levels of the biomarkers in normal and patient serum. Due to the specific binding of antibody to its corresponding antigen, immunosensors based on antibody-antigen interaction are one of the most widely used analytical techniques in the quantitative detection of these diseases. The changed levels of markers in patients are associated with diseases. In this article the biosensors and biomarkers, which were commonly used in terms of monitoring the diagnosis and treatment of cancer and cardiac diseases, are reviewed. In addition, the developed biosensors are compared in terms of precision, reproducibility, regeneration, stability and specificity.


Assuntos
Complexo Antígeno-Anticorpo , Biomarcadores/análise , Doenças Cardiovasculares/diagnóstico , Proteínas dos Microfilamentos/análise , Proteínas de Neoplasias/análise , Neoplasias/diagnóstico , Anticorpos/química , Complexo Antígeno-Anticorpo/química , Antígenos/química , Técnicas Biossensoriais , Doenças Cardiovasculares/metabolismo , Doenças Cardiovasculares/patologia , Técnicas Eletroquímicas , Humanos , Imunoensaio , Limite de Detecção , Nanopartículas Metálicas/química , Proteínas dos Microfilamentos/química , Proteínas de Neoplasias/química , Neoplasias/metabolismo , Neoplasias/patologia , Reprodutibilidade dos Testes
17.
Glia ; 63(4): 684-98, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25530119

RESUMO

Differentiation of oligodendrocyte precursor cells (OPCs) is a prerequisite for both developmental myelination and adult remyelination in the central nervous system. The molecular mechanisms underlying OPC differentiation remain largely unknown. Here, we show that the thirty-kDa HIV-1 Tat interacting protein (TIP30) is a negative regulator in oligodendrocyte development. The TIP30(-/-) mice displayed an increased myelin protein level at postnatal day 14 and 21. By using a primary OPC culture system, we demonstrated that overexpression of TIP30 dramatically inhibited the stage progression of differentiating OPCs, while knockdown of TIP30 enhanced the differentiation of oligodendroglial cells remarkably. Moreover, overexpression of TIP30 was found to sequester the transcription factor Olig1 in the cytoplasm and weaken its nuclear translocation due to the interaction between TIP30 and Olig1, whereas knockdown of TIP30 led to more Olig1 localized in the nucleus in the initiation stage during OPC differentiation. In the cuprizone-induced demyelination model, there was a dramatic increase in NG2-expressing cells with nuclear location of Olig1 in the corpus callosum during remyelination. In contrast, within chronic demyelinated lesions in multiple sclerosis, TIP30 was abnormally expressed in NG2-expressing cells, and few nuclear Olig1 was observed in these cells. Taken together, our findings suggest that TIP30 plays a negative regulatory role in oligodendroglial differentiation.


Assuntos
Diferenciação Celular/genética , Diferenciação Celular/fisiologia , Citoplasma/metabolismo , Oligodendroglia/citologia , Proteínas Repressoras/metabolismo , Células-Tronco/citologia , Proteínas Supressoras de Tumor/metabolismo , Animais , Animais Recém-Nascidos , Antígenos/metabolismo , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Biomarcadores/metabolismo , Técnicas de Cultura de Células , Núcleo Celular/metabolismo , Corpo Caloso/citologia , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Bainha de Mielina/metabolismo , Fibras Nervosas Mielinizadas/metabolismo , Oligodendroglia/metabolismo , Oligodendroglia/fisiologia , Proteoglicanas/metabolismo , Proteínas Repressoras/deficiência , Proteínas Repressoras/genética , Proteínas Supressoras de Tumor/deficiência , Proteínas Supressoras de Tumor/genética
18.
Clin Cancer Res ; 20(21): 5456-67, 2014 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-24893628

RESUMO

PURPOSE: Imiquimod is a Toll-like receptor 7 agonist used topically to treat external genital warts and basal cell carcinoma. We examined the combination of topical imiquimod with intramuscular administration of CRT/E7, a therapeutic human papillomavirus (HPV) vaccine comprised of a naked DNA vector expressing calreticulin fused to HPV16 E7. EXPERIMENTAL DESIGN: Using an orthotopic HPV16 E6/E7(+) syngeneic tumor, TC-1, as a model of high-grade cervical/vaginal/vulvar intraepithelial neoplasia, we assessed if combining CRT/E7 vaccination with cervicovaginal deposition of imiquimod could result in synergistic activities promoting immune-mediated tumor clearance. RESULTS: Imiquimod induced cervicovaginal accumulation of activated E7-specific CD8(+) T cells elicited by CRT/E7 vaccination. Recruitment was not dependent upon the specificity of the activated CD8(+) T cells, but was significantly reduced in mice lacking the IFNγ receptor. Intravaginal imiquimod deposition induced upregulation of CXCL9 and CXCL10 mRNA expression in the genital tract, which are produced in response to IFNγ receptor signaling and attract cells expressing their ligand, CXCR3. The T cells attracted by imiquimod to the cervicovaginal tract expressed CXCR3 as well as CD49a, an integrin involved in homing and retention of CD8(+) T cells at mucosal sites. Our results indicate that intramuscular CRT/E7 vaccination in conjunction with intravaginal imiquimod deposition recruits antigen-specific CXCR3(+) CD8(+) T cells to the genital tract. CONCLUSIONS: Several therapeutic HPV vaccination clinical trials using a spectrum of DNA vaccines, including vaccination in concert with cervical imiquimod, are ongoing. Our study identifies a mechanism by which these strategies could provide therapeutic benefit. Our findings support accumulating evidence that manipulation of the tumor microenvironment can enhance the therapeutic efficacy of strategies that induce tumor-specific T cells.


Assuntos
Aminoquinolinas/farmacologia , Antígenos/imunologia , Linfócitos T CD8-Positivos/efeitos dos fármacos , Linfócitos T CD8-Positivos/imunologia , Genitália/efeitos dos fármacos , Interferon gama/imunologia , Receptores Toll-Like/agonistas , Animais , Feminino , Genitália/virologia , Imiquimode , Integrina alfa1/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Neoplasias/tratamento farmacológico , Neoplasias/imunologia , Neoplasias/virologia , Proteínas Oncogênicas Virais/imunologia , Proteínas E7 de Papillomavirus/imunologia , Vacinas contra Papillomavirus/imunologia , Receptores de Interferon/imunologia , Proteínas Repressoras/imunologia , Vacinação/métodos , Vacinas de DNA/imunologia
19.
Am J Physiol Cell Physiol ; 307(1): C25-38, 2014 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-24788248

RESUMO

Tissue growth and function depend on vascularization, and vascular insufficiency or excess exacerbates many human diseases. Identification of the biological processes involved in angiogenesis will dictate strategies to modulate reduced or excessive vessel formation. We examine the essential role of pericytes. Their heterogeneous morphology, distribution, origins, and physiology have been described. Using double-transgenic Nestin-GFP/NG2-DsRed mice, we identified two pericyte subsets. We found that Nestin-GFP(-)/NG2-DsRed(+) (type-1) and Nestin-GFP(+)/NG2-DsRed(+) (type-2) pericytes attach to the walls of small and large blood vessels in vivo; in vitro, type-2, but not type-1, pericytes spark endothelial cells to form new vessels. Matrigel assay showed that only type-2 pericytes participate in normal angiogenesis. Moreover, when cancer cells were transplanted into Nestin-GFP/NG2-DsRed mice, type-1 pericytes did not penetrate the tumor, while type-2 pericytes were recruited during its angiogenesis. As inhibition of angiogenesis is a promising strategy in cancer therapy, type-2 pericytes may provide a cellular target susceptible to signaling and pharmacological manipulation in treating malignancy. This work also reports the potential of type-2 pericytes to improve blood perfusion in ischemic hindlimbs, indicating their potential for treating ischemic illnesses.


Assuntos
Neoplasias Encefálicas/irrigação sanguínea , Glioblastoma/irrigação sanguínea , Isquemia/fisiopatologia , Músculo Esquelético/irrigação sanguínea , Neovascularização Patológica , Neovascularização Fisiológica , Pericitos/patologia , Actinas/genética , Animais , Antígenos/genética , Biomarcadores/metabolismo , Células Cultivadas , Técnicas de Cocultura , Modelos Animais de Doenças , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Membro Posterior , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Isquemia/metabolismo , Isquemia/patologia , Proteínas Luminescentes/genética , Proteínas Luminescentes/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Nus , Camundongos Transgênicos , Nestina/genética , Pericitos/metabolismo , Fenótipo , Regiões Promotoras Genéticas , Proteoglicanas/genética , Fatores de Tempo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA