Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
Nucleic Acid Ther ; 32(6): 486-496, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-35895511

RESUMO

Metabolic syndrome (MetS) is a pathological condition characterized by abdominal obesity, insulin resistance, hypertension, and hyperlipidemia. Sirtuin 1 (SIRT1), a highly conserved histone deacetylase, is characterized as a key metabolic regulator and protector against aging-associated pathologies, including MetS. In this study, we investigate the therapeutic potential of activating SIRT1 using small activating RNAs (saRNA), thereby reducing inflammatory-like responses and re-establishing normal lipid metabolism. SIRT1 saRNA significantly increased SIRT1 messenger RNA (mRNA) and protein levels in both lipopolysaccharide-stimulated and nonstimulated macrophages. SIRT1 saRNA significantly decreased inflammatory-like responses, by reducing mRNA levels of key inflammatory cytokines, such as Tumor Necrosis Factor alpha, Interleukin 1 beta (IL-1ß), Interleukin 6 (IL-6), and chemokines Monocyte Chemoattractant Protein-1 and keratinocyte chemoattractant. SIRT1 overexpression also significantly reduced phosphorylation of nuclear factor-κB and c-Jun N-terminal kinase, both key signaling molecules for the inflammatory pathway. To investigate the therapeutic effect of SIRT1 upregulation, we treated a high-fat diet model with SIRT1 saRNA conjugated to a transferrin receptor aptamer for delivery to the liver and cellular internalization. Animals in the SIRT1 saRNA treatment arm demonstrated significantly decreased weight gain with a significant reduction in white adipose tissue, triglycerides, fasting glucose levels, and intracellular lipid accumulation. These suggest treatment-induced changes to lipid and glucose metabolism in the animals. The results of this study demonstrate that targeted activation of SIRT1 by saRNAs is a potential strategy to reverse MetS.


Assuntos
Síndrome Metabólica , Humanos , Síndrome Metabólica/genética , Síndrome Metabólica/terapia , RNA Mensageiro , Expressão Gênica , Lipídeos , Sirtuína 1/genética
2.
Transl Oncol ; 13(3): 100749, 2020 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-32143178

RESUMO

Discovery of biomarkers is critical to understand tumor heterogeneity and microenvironment. To determine differently expressed makers on cancer tissue for comprehensive profiling, the multiplexed tissue imaging mass cytometer (IMC) which uniquely combines time-of-flight mass spectrometry with metal-labeling technology to enable breakthrough discovery on single cell level was employed to investigate the expression of seven markers related to the epithelial-to-mesenchymal transition [α-smooth muscle actin (α-SMA), vimentin, collagen I, cytokeratin 7, pan-keratin], tumor proliferation (Ki-67), and human leucocyte antigen (HLA-DR) on human pancreatic cancer tissue. The difference was analyzed using bioinformatic tools. We observed the high expression of α-SMA, vimentin, collagen I, and Ki-67 on grade I but not on grade III. HLA-DR was highly expressed on grade I/III but not on grade II. Overall, the expression of markers has elucidated the heterogeneity intratumors. Additionally, to identify biomarkers on pancreatic cancer cells by blind systematic evolution of ligands by exponential enrichment (SELEX), aptamer pull-down assay and liquid chromatography-tandem mass spectrometry were used. Mortalin was identified as a potential a prognostic marker of pancreatic cancer. Our studies demonstrate that the IMC and blind SELEX might be implemented to discover biomarkers which can be used to better understand tumor biology and biomedical research applications.

3.
Mol Ther Nucleic Acids ; 18: 142-154, 2019 Dec 06.
Artigo em Inglês | MEDLINE | ID: mdl-31546149

RESUMO

Pancreatic ductal adenocarcinoma (PDAC) is one of the most aggressive malignancies; it preferentially metastasizes to the liver and is the main cause of death from this disease. In previous studies, small activating RNA against CCAAT/enhancer-binding protein-α (C/EBPα-saRNA) demonstrated efficacy of PDAC in a local subcutaneous tumor model. In this study, we focused on the efficacy of C/EBPα-saRNA in advanced stage PDAC. For targeted delivery, we selected a new anti-transferrin receptor aptamer (TR14), which demonstrated a high binding affinity to target proteins. The TR14 aptamer was internalized with clathrin-mediated endocytosis, distributed in early endosome, late endosome, and lysosome subcellularly. To investigate its anti-tumor effects to advanced PDAC, we conjugated C/EBPα-saRNA to TR14. Treatment of pancreatic cancer cells with the conjugates upregulated expression of C/EBPα and its downstream target p21, and inhibited cell proliferation. For in vivo assays, we established an advanced PDAC mouse model by engrafting luciferase reporter-PANC-1 cells directly into the livers of non-obese diabetic/severe combined immunodeficiency (NOD/SCID) mice. After treatment of aptamer-C/EBPα conjugates, we observed significant reduction of tumor growth in this advanced PDAC mouse model. Combinational treatment of the conjugates with gemcitabine also demonstrated enhanced anti-tumor effects in advanced PDAC. This suggests that aptamer-C/EBPα conjugates could be used as an adjuvant, along with other conventional anti-cancer drugs in advanced PDAC. In conclusion, targeted delivery of C/EBPα-saRNAs by aptamers might have potential therapeutic effects in advanced PDAC.

4.
Mol Ther Nucleic Acids ; 14: 131-141, 2019 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-30594071

RESUMO

Human glioblastoma (GBM) is the most aggressive malignancy of the CNS, with less than 5% survival. Despite great efforts to find effective therapeutics, current options remain very limited. To develop a targeted cancer therapeutic, we selected RNA aptamers against platelet-derived growth factor receptor α (PDGFRα), which is a receptor tyrosine kinase. One RNA aptamer (PDR3) with high affinity (0.25 nM) showed PDGFRα specificity and was internalized in U251-MG cells. Following treatment with the PDR3 aptamer, expression of the transcription factor STAT3 (signal transducer and activator of transcription 3) was inhibited, whereas the expression of the histone demethylase JMJD3 and the tumor suppressor p53 were upregulated. PDR3 also upregulated serine phosphorylation of p53, which subsequently mediated apoptosis through the death receptors: tumor necrosis factor (TNF)-related apoptosis-inducing ligand receptors 1/2 (TRAIL-R1/R2), Fas-associated via death domain (FADD), and Fas. PDR3 significantly decreased cell viability in a dose-dependent manner. Furthermore, translocation of PDR3 into the nucleus induced hypomethylation at the promoters of cyclin D2. To assess the feasibility of targeted delivery, we conjugated PDR3 aptamer with STAT3-siRNA for a chimera. The PDR3-siSTAT3 chimera successfully inhibited the expression of target genes and showed significant inhibition of cell viability. In summary, our results show that well-tailored RNA aptamers targeting the PDGFRα-STAT3 axis have the potential to act as anti-cancer therapeutics in GBM.

5.
Adv Drug Deliv Rev ; 134: 22-35, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-29981799

RESUMO

The structural flexibility and small size of aptamers enable precise recognition of cellular elements for imaging and therapeutic applications. The process by which aptamers are taken into cells depends on their targets but is typically clathrin-mediated endocytosis or macropinocytosis. After internalization, most aptamers are transported to endosomes, lysosomes, endoplasmic reticulum, Golgi apparatus, and occasionally mitochondria and autophagosomes. Intracellular aptamers, or "intramers," have versatile functions ranging from intracellular RNA imaging, gene regulation, and therapeutics to allosteric modulation, which we discuss in this review. Immune responses to therapeutic aptamers and the effects of G-quadruplex structure on aptamer function are also discussed.


Assuntos
Aptâmeros de Nucleotídeos/metabolismo , Aptâmeros de Nucleotídeos/farmacocinética , Aptâmeros de Nucleotídeos/química , Aptâmeros de Nucleotídeos/farmacologia , Avaliação Pré-Clínica de Medicamentos , Transferência Ressonante de Energia de Fluorescência , Humanos , Imagem Óptica , Fatores de Transcrição/antagonistas & inibidores , Fatores de Transcrição/metabolismo
6.
Pharmaceuticals (Basel) ; 11(3)2018 Jul 19.
Artigo em Inglês | MEDLINE | ID: mdl-30029472

RESUMO

Imaging is not only seeing, but also believing. For targeted imaging modalities, nucleic acid aptamers have features such as superior recognition of structural epitopes and quick uptake in target cells. This explains the emergence of an evolved new class of aptamers into a wide spectrum of imaging applications over the last decade. Genetically encoded biosensors tagged with fluorescent RNA aptamers have been developed as intracellular imaging tools to understand cellular signaling and physiology in live cells. Cancer-specific aptamers labeled with fluorescence have been used for assessment of clinical tissue specimens. Aptamers conjugated with gold nanoparticles have been employed to develop innovative mass spectrometry tissue imaging. Also, use of chemically conjugated cancer-specific aptamers as probes for non-invasive and high-resolution imaging has been transformative for in vivo imaging in multiple cancers.

7.
Curr Pharm Biotechnol ; 19(8): 604-610, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29804529

RESUMO

BACKGROUND: RNA is increasingly recognized as a powerful molecule that can be used to control gene expression. Sophisticated, well-engineered RNA-based regulators are being developed as oligotherapeutics. METHODS: In particular, small activating RNAs (saRNAs) are promising therapeutic options for targeting human diseases. Numerous saRNAs targeting multiple cancers have been developed in preclinical models. One saRNA targeting C/EBPα is currently undergoing clinical trials in liver cancer. RESULTS AND CONCLUSION: In this review, we describe the current working model of the intracellular mechanism of saRNA, discuss the recent progress of saRNA therapeutics in preclinical and clinical trials, and current advances in targeted delivery using aptamers in detail.


Assuntos
Marcação de Genes/métodos , Neoplasias/genética , Neoplasias/terapia , RNA Interferente Pequeno/administração & dosagem , RNA Interferente Pequeno/genética , Ensaios Clínicos como Assunto/métodos , Expressão Gênica , Marcação de Genes/tendências , Terapia Genética/métodos , Terapia Genética/tendências , Humanos
8.
Curr Opin Oncol ; 29(5): 366-374, 2017 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-28692589

RESUMO

PURPOSE OF REVIEW: We will describe recently discovered smart aptamers with tumor specificity, with an emphasis on targeted delivery of novel therapeutic molecules, cancer-specific biomarkers, and immunotherapy. RECENT FINDINGS: The development of cancer-specific aptamers has facilitated targeted delivery of potent therapeutic molecules to cancer cells without harming nontumoral cells. This specificity also makes it possible to discover novel cancer biomarkers. Furthermore, alternative immune-checkpoint blockade aptamers have been developed for combinational immunotherapy. SUMMARY: Aptamers selected against cancer cells show cancer specificity, which has great potential for targeting. First, functionalizing targeted aptamers with therapeutic molecule payloads (e.g., small activating RNAs, antimitotic drugs, therapeutic antibodies, and peptides) facilitates successful delivery into cancer cells. This approach greatly improves the therapeutic index by minimizing side-effects in nontumoral cells. Second, cancer-specific proteins have been identified as cancer biomarkers through in-vitro and in-vivo selection, aptamer pull-down assays, and mass spectrometry. These newly discovered biomarkers improve therapeutic intervention and diagnostic specificity. In addition, the development of alternative immune-checkpoint blockade aptamers is suggested for use in combinational immunotherapeutic with current immune blockade regimens, to reduce the resistance and exhaustion of T cells in clinical trials. VIDEO ABSTRACT: http://links.lww.com/COON/A21.


Assuntos
Aptâmeros de Nucleotídeos/uso terapêutico , Aptâmeros de Peptídeos/uso terapêutico , Neoplasias/tratamento farmacológico , Aptâmeros de Nucleotídeos/genética , Aptâmeros de Nucleotídeos/farmacocinética , Aptâmeros de Peptídeos/genética , Aptâmeros de Peptídeos/farmacocinética , Humanos , Terapia de Alvo Molecular , Neoplasias/genética , Neoplasias/metabolismo
9.
Adv Exp Med Biol ; 983: 173-188, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28639199

RESUMO

Pancreatic cancer is estimated to become the second-leading cause of cancer-related mortality by 2020. While the death rates of most other cancers continue to decline recently, the death rates of pancreatic cancer are still increasing, with less than 5% of patients achieving 5-year survival. Despite great efforts to improve treatment with combinational therapies in pancreatic cancer patients, limited progress has been made. V-Ki-ras2 Kirsten rat sarcoma viral oncogene homolog (KRAS) has been depicted as a therapeutic target in pancreatic cancer for many years. However, the clinical outcome of KRAS-directed therapies has not been successful, suggesting that KRAS is an undruggable target. For the new druggable target, epigenetically silenced transcriptional factor C/EBPα (CCAAT/enhancer-binding protein α), upregulator of a strong inhibitor of cell proliferation (p21), is upregulated by small activating RNA (saRNA) in pancreatic cancer. For the cell type-specific delivery, pancreatic cancer-specific 2'-Fluoropyrimidine RNA-aptamers (2'F-RNAs) are conjugated with C/EBPα-saRNA via sticky bridge sequences. The conjugates of aptamer-C/EBPα-saRNA upregulate the expression of C/EBPα in vitro and inhibit the tumor growth in vivo. It suggests that aptamer-mediated targeted delivery of therapeutic C/EBPα-saRNA might be the effective therapeutics under the current therapeutic modality failure in pancreatic cancer.


Assuntos
Aptâmeros de Nucleotídeos , Proteína alfa Estimuladora de Ligação a CCAAT/uso terapêutico , Neoplasias Pancreáticas/terapia , RNA/uso terapêutico , Proliferação de Células , Humanos , Quinases Ativadas por p21
10.
Mol Cancer Res ; 15(7): 811-820, 2017 07.
Artigo em Inglês | MEDLINE | ID: mdl-28396463

RESUMO

Identifying targets that are exposed on the plasma membrane of tumor cells, but expressed internally in normal cells, is a fundamental issue for improving the specificity and efficacy of anticancer therpeutics. Using blind cell Systemic Evolution of Ligands by EXponetial enrichment (SELEX), which is untargeted SELEX, we have identified an aptamer, P15, which specifically bound to the human pancreatic adenocarcinoma cells. To identify the aptamer binding plasma membrane protein, liquid chromatography tandem mass spectrometry (LC-MS/MS) was used. The results of this unbiased proteomic mass spectrometry approach identified the target of P15 as the intermediate filament vimentin, biomarker of epithelial-mesenchymal transition (EMT), which is an intracellular protein but is specifically expressed on the plasma membrane of cancer cells. As EMT plays a pivotal role to transit cancer cells to invasive cells, tumor cell metastasis assays were performed in vitro P15-treated pancreatic cancer cells showed the significant inhibition of tumor metastasis. To investigate the downstream effects of P15, EMT-related gene expression analysis was performed to identify differently expressed genes (DEG). Among five DEGs, P15-treated cells showed the downregulated expression of matrix metallopeptidase 3 (MMP3), which is involved in cancer invasion. These results, for the first time, demonstrate that P15 binding to cell surface vimentin inhibits the tumor cell invasion and is associated with reduced MMP3 expression. Thus, suggesting that P15 has potential as an anti-metastatic therapy in pancreatic cancer.Implications: This study reveals that anti-vimentin RNA aptamers selected via blind-SELEX inhibit the tumor cell metastasis. Mol Cancer Res; 15(7); 811-20. ©2017 AACR.


Assuntos
Adenocarcinoma/genética , Aptâmeros de Nucleotídeos/administração & dosagem , Biomarcadores Tumorais/genética , Metaloproteinase 3 da Matriz/genética , Neoplasias Pancreáticas/genética , Adenocarcinoma/patologia , Aptâmeros de Nucleotídeos/genética , Linhagem Celular Tumoral , Transição Epitelial-Mesenquimal/genética , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Ligantes , Invasividade Neoplásica/genética , Invasividade Neoplásica/patologia , Metástase Neoplásica , Neoplasias Pancreáticas/patologia , Proteômica , Técnica de Seleção de Aptâmeros
12.
Mol Ther Nucleic Acids ; 6: 80-88, 2017 Mar 17.
Artigo em Inglês | MEDLINE | ID: mdl-28325302

RESUMO

Aptamer-drug conjugates (ApDCs) have the potential to improve the therapeutic index of traditional chemotherapeutic agents due to their ability to deliver cytotoxic drugs specifically to cancer cells while sparing normal cells. This study reports on the conjugation of cytotoxic drugs to an aptamer previously described by our group, the pancreatic cancer RNA aptamer P19. To this end, P19 was incorporated with gemcitabine and 5-fluorouracil (5-FU), or conjugated to monomethyl auristatin E (MMAE) and derivative of maytansine 1 (DM1). The ApDCs P19-dFdCMP and P19-5FdUMP were shown to induce the phosphorylation of histone H2AX on Ser139 (γ-H2AX) and significantly inhibited cell proliferation by 51%-53% in PANC-1 and by 54%-34% in the gemcitabine-resistant pancreatic cancer cell line AsPC-1 (p ≤ 0.0001). P19-MMAE and P19-DM1 caused mitotic G2/M phase arrest and inhibited cell proliferation by up to 56% in a dose-dependent manner when compared to the control group (p ≤ 0.001). In addition, the cytotoxicity of P19-MMAE and P19-DM1 in normal cells and the control human breast cancer cell line MCF7 was minimal. These results suggest that this approach may be useful in decreasing cytotoxic side effects in non-tumoral tissue.

13.
Mol Ther ; 24(6): 1106-1116, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-26983359

RESUMO

The 5-year survival rate for pancreatic ductal adenocarcinoma (PDAC) remains dismal despite current chemotherapeutic agents and inhibitors of molecular targets. As the incidence of PDAC constantly increases, more effective multidrug approaches must be made. Here, we report a novel method of delivering antitumorigenic therapy in PDAC by upregulating the transcriptional factor CCAAT/enhancer-binding protein-α (C/EBPα), recognized for its antiproliferative effects. Small activating RNA (saRNA) duplexes designed to increase C/EBPα expression were linked onto PDAC-specific 2'-Fluropyrimidine RNA aptamers (2'F-RNA) - P19 and P1 for construction of a cell type-specific delivery vehicle. Both P19- and P1-C/EBPα-saRNA conjugates increased expression of C/EBPα and significantly suppressed cell proliferation. Tail vein injection of the saRNA/aptamer conjugates in PANC-1 and in gemcitabine-resistant AsPC-1 mouse-xenografts led to reduced tumor size with no observed toxicity. To exploit the specificity of the P19/P1 aptamers for PDAC cells, we also assessed if conjugation with Cy3 would allow it to be used as a diagnostic tool on archival human pancreatic duodenectomy tissue sections. Scoring pattern from 72 patients suggested a positive correlation between high fluorescent signal in the high mortality patient groups. We propose a novel aptamer-based strategy for delivery of targeted molecular therapy in advanced PDAC where current modalities fail.


Assuntos
Aptâmeros de Nucleotídeos/administração & dosagem , Proteína alfa Estimuladora de Ligação a CCAAT/genética , Carcinoma Ductal Pancreático/terapia , Neoplasias Pancreáticas/terapia , RNA/administração & dosagem , Animais , Aptâmeros de Nucleotídeos/farmacologia , Carcinoma Ductal Pancreático/genética , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos , Humanos , Camundongos , Especificidade de Órgãos , Neoplasias Pancreáticas/genética , RNA/farmacologia , Resultado do Tratamento , Regulação para Cima , Ensaios Antitumorais Modelo de Xenoenxerto
14.
FEMS Immunol Med Microbiol ; 61(3): 359-63, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21208291

RESUMO

Swine hepatitis E virus (sHEV) has been discovered to be almost ubiquitous in pigs, and is antigenically and genetically related to human HEV. Proteomic analysis was used to identify altered protein expression in swine liver, using two-dimensional electrophoresis and peptide mass fingerprinting. A total of 10 protein spots exhibited significant alterations in the sHEV-infected organ. The upregulation of apolipoprotein E (Apo E) and downregulation of ferritin heavy chain were confirmed by Western analysis and by semi-quantitative reverse transcription-PCR. The elevated expression of Apo E may provide a novel insight into molecular responses to HEV infection in swine.


Assuntos
Apoferritinas/análise , Apolipoproteínas/análise , Hepatite E/veterinária , Fígado/patologia , Proteoma/análise , Doenças dos Suínos/patologia , Animais , Western Blotting , Regulação para Baixo , Hepatite E/patologia , Hepatite E/virologia , Vírus da Hepatite E/patogenicidade , Humanos , Fígado/virologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Suínos , Doenças dos Suínos/virologia , Regulação para Cima
15.
J Vet Med Sci ; 73(2): 275-7, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-20953132

RESUMO

Clinical grape poisoning in two dogs (a 1.6-year-old male Shih Tzu and a 5-year-old female Yorkshire Terrier) was described in the present study. Clinical signs included decreased urine output in the Shih Tzu and ataxia in the Yorkshire Terrier after grape ingestion. The Shih Tzu died 5 days post-grape ingestion, while the Yorkshire Terrier died 3 days post-grape ingestion. Erythematous serosae and mucosae, multifocal red small intestinal foci, and blood and grape seeds were identified in the intestinal lumen. Brownish-yellow crystals were bilaterally identified in the renal pelvis. The primary histological findings were acute tubular necrosis of the proximal convoluted tubules, severe necrosis, and mineralization in the renal cortical tubules. Blood urea nitrogen, creatinine, and alanine aminotransferase were increased in the dogs. Many Korean veterinary clinicians have suspected clinical grape poisoning. However, to our knowledge, grape poisoning has not been identified by pathologic and clinicopathologic basis until this writing in Korea. Education and knowledge about the risks of grape poisoning is necessary for the prevention of accidental exposures.


Assuntos
Doenças do Cão/etiologia , Nefropatias/veterinária , Vitis/intoxicação , Animais , Cães , Evolução Fatal , Feminino , Histocitoquímica , Nefropatias/etiologia , Masculino
16.
Antiviral Res ; 88(1): 19-24, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20615431

RESUMO

Porcine circovirus type 2 (PCV2) is the main causative agent of porcine circovirus-associated diseases (PCVD), which is responsible for economic losses in the swine industry. The capsid protein of PCV2 has important role for virus neutralization that blocks viral infection. To develop the therapeutic agents, two 2'F-RNA aptamers that bound to the PCV2 capsid protein with nanomole affinity were isolated from a 2'F-RNA library by the Systematic Evolution of Ligands by EXponential enrichment (SELEX). The binding affinity of aptamers was analyzed by Electrophoretic Mobility shift assay (EMSA) and surface plasmon resonance (SPR) analysis. The RNA aptamers have been shown to exhibit high affinity and specificity to PCV2 capsid protein and to neutralize PCV2 infectivity in PK-15 cells in dose dependent manner. Neutralizing aptamers such as this could be promising candidates in developing efficacious anti-PCV2 drugs as well as therapeutic delivery reagent.


Assuntos
Antivirais/farmacologia , Aptâmeros de Nucleotídeos/metabolismo , Aptâmeros de Nucleotídeos/farmacologia , Proteínas do Capsídeo/metabolismo , Circovirus/efeitos dos fármacos , Animais , Aptâmeros de Nucleotídeos/uso terapêutico , Capsídeo/efeitos dos fármacos , Proteínas do Capsídeo/genética , Linhagem Celular , Circovirus/genética , Circovirus/fisiologia , Técnica Indireta de Fluorescência para Anticorpo , Expressão Gênica , Reação em Cadeia da Polimerase , Pirimidinas , RNA , Proteínas Recombinantes , Análise de Sequência de DNA , Suínos , Virulência/efeitos dos fármacos , Replicação Viral/efeitos dos fármacos
17.
J Gen Virol ; 91(Pt 10): 2585-91, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20573857

RESUMO

Porcine circovirus type 2 (PCV2) is the main causative agent of porcine circovirus-associated disease, such as post-weaning multisystemic wasting syndrome, which involves lymphocyte depletion. However, little is known about the molecular mechanisms of lymphoid depletion. To gain insight into the interaction between virus and host cells, microarrays were used to analyse changes in genomic expression in lymph nodes following PCV2 infection of pigs, together with negative controls. Total RNA was subjected to microarray analysis with an Affymetrix Porcine Genome Array GeneChip. Of the 23,256 pig genes arrayed on a chip, 160 genes showed altered expression after infection (upregulated, 64; downregulated, 96). The altered genomic expression of 18 selected genes was confirmed by quantitative real-time PCR. The expression changes of numerous genes involved in innate immune defence (TLR1, CD14 and CD180), immunosuppressed responses (FGL2 and GPNMB), pro-inflammatory signals (galectin-3) and fasting processes (ANGPTL-4) indicate that PCV2 has developed an intricate mechanism to cause immunosuppression, inflammatory cell infiltration and weight loss in pigs. The results of this study provide a basis for understanding the molecular pathogenesis of PCV2 infection.


Assuntos
Infecções por Circoviridae/veterinária , Circovirus/patogenicidade , Perfilação da Expressão Gênica , Linfonodos/imunologia , Linfonodos/virologia , Doenças dos Suínos/imunologia , Doenças dos Suínos/virologia , Animais , Infecções por Circoviridae/imunologia , Infecções por Circoviridae/virologia , Interações Hospedeiro-Patógeno , Tolerância Imunológica , Linfócitos/imunologia , Análise de Sequência com Séries de Oligonucleotídeos , Reação em Cadeia da Polimerase , Suínos
18.
J Virol ; 84(16): 8241-9, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20534854

RESUMO

We previously showed that agonistic antibodies to CD40 could substitute for CD4 T-cell help and prevent reactivation of murine gammaherpesvirus 68 (MHV-68) in the lungs of major histocompatibility complex (MHC) class II(-/-) (CII(-/-)) mice, which are CD4 T cell deficient. Although CD8 T cells were required for this effect, no change in their activity was detected in vitro. A key question was whether anti-CD40 treatment (or CD4 T-cell help) changed the function of CD8 T cells or another cell type in vivo. To address this question, in the present study, we showed that adoptive transfer of CD8 T cells from virus-infected wild-type mice or anti-CD40-treated CII(-/-) mice caused a significant reduction in lung viral titers, in contrast to those from control CII(-/-) mice. Anti-CD40 treatment also greatly prolonged survival of infected CII(-/-) mice. This confirms that costimulatory signals cause a change in CD8 T cells enabling them to maintain effective long-term control of MHV-68. We investigated the nature of this change and found that expression of the inhibitory receptor PD-1 was significantly increased on CD8 T cells in the lungs of MHV-68-infected CII(-/-), CD40(-/-), or CD80/86(-/-) mice, compared with that in wild-type or CD28/CTLA4(-/-) mice, correlating with the level of viral reactivation. Furthermore, blocking PD-1-PD-L1 interactions significantly reduced viral reactivation in CD4 T-cell-deficient mice. In contrast, the absence of another inhibitory receptor, NKG2A, had no effect. These data suggest that CD4 T-cell help programs a change in CD8 T-cell function mediated by altered PD-1 expression, which enables effective long-term control of MHV-68.


Assuntos
Antígenos de Diferenciação/metabolismo , Antígeno B7-1/metabolismo , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Glicoproteínas de Membrana/metabolismo , Peptídeos/metabolismo , Rhadinovirus/imunologia , Transferência Adotiva , Animais , Antígeno B7-H1 , Antígenos CD40/antagonistas & inibidores , Feminino , Antígenos de Histocompatibilidade Classe II/genética , Pulmão/virologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptor de Morte Celular Programada 1 , Ligação Proteica , Análise de Sobrevida
19.
J Vet Med Sci ; 72(9): 1195-202, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20453453

RESUMO

Maesil (Prunus mume) has long been used as a traditional drug and healthy food in East Asian countries. It possesses a number of beneficial biological activities including potential antimicrobial effects against pathogens. Probiotics also have antibacterial effects. Moreover, some probiotics have an important role in regulating the immune system. The present study evaluated the immune enhancing effects of fermented Maesil with probiotics (Saccharomyces cerevisiae, Bacillus subtilis and Lactobacillus acidophilus) in mice, especially against Bordetella bronchiseptica, as an initial step towards the development of feed supplements for the promotion of immune activity and prevention of disease, especially in pigs. Continuous ingestion of fermented Maesil with probiotics markedly increased the macrophage ratio in peripheral blood and the T lymphocyte ratio in the spleen. In addition, antibody production against formalin-killed B. bronchiseptica significantly increased in the mice fed fermented Maesil compared with the control group. The number of leukocytes was significantly higher in the bronchio-alveolar lavage obtained from the fermented Maesil-fed animals compared to it in the control group at day 3 (maximal peak time) after experimental B. bronchiseptica infection. Moreover, at 7 day post-infection, relative messenger RNA expression levels of tumor necrosis factor- α and interferon-γ were significantly increased in splenocytes of mice fed fermented Maesil compared with those in the control group. Taken together, these findings suggest that feed containing fermented Maesil with probiotics enhances immune activity in mice, especially against B. bronchiseptica, via the potent stimulation of non-specific immune responses.


Assuntos
Bordetella bronchiseptica/imunologia , Prunus/imunologia , Actinas/genética , Animais , Infecções por Bordetella/sangue , Infecções por Bordetella/imunologia , Infecções por Bordetella/veterinária , Bordetella bronchiseptica/efeitos dos fármacos , Citocinas/genética , Primers do DNA , Suplementos Nutricionais , Fermentação , Interferon gama/genética , Contagem de Leucócitos/veterinária , Camundongos , Probióticos/farmacologia , Probióticos/uso terapêutico , Fator de Necrose Tumoral alfa/genética , Medicina Veterinária
20.
Am J Pathol ; 176(2): 800-11, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-20042666

RESUMO

Secondary bacterial infections that follow infection with influenza virus result in considerable morbidity and mortality in young children, the elderly, and immunocompromised individuals and may also significantly increase mortality in normal healthy adults during influenza pandemics. We herein describe a mouse model for investigating the interaction between influenza virus and the bacterium Haemophilus influenzae. Sequential infection with sublethal doses of influenza and H. influenzae resulted in synergy between the two pathogens and caused mortality in immunocompetent adult wild-type mice. Lethality was dependent on the interval between administration of the bacteria and virus, and bacterial growth was prolonged in the lungs of dual-infected mice, although influenza virus titers were unaffected. Dual infection induced severe damage to the airway epithelium and confluent pneumonia, similar to that observed in victims of the 1918 global influenza pandemic. Increased bronchial epithelial cell death was observed as early as 1 day after bacterial inoculation in the dual-infected mice. Studies using knockout mice indicated that lethality occurs via a mechanism that is not dependent on Fas, CCR2, CXCR3, interleukin-6, tumor necrosis factor, or Toll-like receptor-4 and does not require T or B cells. This model suggests that infection with virulent strains of influenza may predispose even immunocompetent individuals to severe illness on secondary infection with H. influenzae by a mechanism that involves innate immunity, but does not require tumor necrosis factor, interleukin-6, or signaling via Toll-like receptor-4.


Assuntos
Modelos Animais de Doenças , Infecções por Haemophilus/mortalidade , Haemophilus influenzae/fisiologia , Vírus da Influenza A/fisiologia , Infecções por Orthomyxoviridae/mortalidade , Imunidade Adaptativa/fisiologia , Animais , Células Cultivadas , Cães , Infecções por Haemophilus/complicações , Infecções por Haemophilus/patologia , Infecções por Haemophilus/virologia , Humanos , Pulmão/patologia , Pulmão/virologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Infecções por Orthomyxoviridae/complicações , Infecções por Orthomyxoviridae/patologia , Infecções por Orthomyxoviridae/virologia , Superinfecção/imunologia , Superinfecção/mortalidade , Superinfecção/patologia , Superinfecção/virologia , Carga Viral
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA