Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Br J Cancer ; 122(7): 971-977, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-32063605

RESUMEN

BACKGROUND: TG01 is the first cancer immunotherapy targeting KRAS oncogenic mutations. This study assessed the safety and efficacy of TG01/GM-CSF in patients with resected pancreatic adenocarcinoma. METHODS: Patients with stage I or II pancreatic adenocarcinoma who had undergone surgical resection (R0 or R1) received adjuvant gemcitabine with TG01/GM-CSF using two schedules of vaccination. Immune response was defined as a positive delayed-type hypersensitivity (DTH) response and/or positive T-cell proliferation assay. RESULTS: Thirty-two patients were enrolled between February 2013 and May 2016. Nineteen were treated with the high antigen burden, with four serious adverse reactions considered possibly related to TG01 treatment, including three allergic reactions. On this basis, a further 13 patients received a modified vaccination schedule with reduced antigen burden, with no serious adverse events related to TG01. Ninety-five percent patients in the main cohort and 92% in the modified cohort had a positive immune response. Median overall survival (OS) was 33.1 months, and median disease-free survival (DFS) was 13.9 months for the main cohort. For the modified cohort, the median OS was 34.3 months and median DFS was 19.5 months. CONCLUSIONS: TG01/GM-CSF with gemcitabine was well tolerated, with high levels of immune activation. OS and DFS compare favourably with published data for adjuvant gemcitabine. CLINICAL TRIAL REGISTRATION: This clinical trial was registered at ClinicalTrials.gov (NCT02261714).


Asunto(s)
Adenocarcinoma/tratamiento farmacológico , Desoxicitidina/análogos & derivados , Factor Estimulante de Colonias de Granulocitos y Macrófagos/uso terapéutico , Neoplasias Pancreáticas/tratamiento farmacológico , Adenocarcinoma/patología , Anciano , Desoxicitidina/farmacología , Desoxicitidina/uso terapéutico , Femenino , Factor Estimulante de Colonias de Granulocitos y Macrófagos/farmacología , Humanos , Masculino , Persona de Mediana Edad , Neoplasias Pancreáticas/patología , Neoplasias Pancreáticas/cirugía , Gemcitabina , Neoplasias Pancreáticas
2.
J Med Virol ; 92(8): 1309-1315, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-31944306

RESUMEN

Despite new therapies, the estimated 229 875 women living with ovarian cancer have a 5-year survival rate of 47.6%. This cavity-localized cancer lends itself to local administration of modalities, such as the oncolytic adenovirus (Ad) Ad5/3-D24-granulocyte-macrophage colony-stimulating factor virus (ONCOS-102). Its repeated administration to a patient with chemotherapy-refractory ovarian cancer induced CD8+ antitumor immune responses with the overall survival reaching 40 months. Here we probe the dominant receptor used by ONCOS-102 in four established epithelial ovarian cancer cell lines. Ad3 can use the desmoglein-2 (DSG2) and CD46 receptors on susceptible cells. DSG2 was nearly absent in A2780 cells but was expressed in more than 90% of OAW42, OVCAR3, and OV-90 cells. After 96 hours, ONCOS-102 treatment showed significant oncolytic activity (≧50%) in OAW42, OVCAR3, and OV-90 cells, but minimal activity in A2780 cells, suggesting DSG2 as the dominant receptor for ONCOS-102. Furthermore, retrospective analyses of phase I clinical trial of ONCOS-102 treatment of 12 patients with varied tumors indicated a correlation between viral genomes in blood and DSG2 RNA expression. These data support the role of DSG2 expression on cancer cells in virus infectivity and the continued development of ONCOS-102 for ovarian cancer treatment.


Asunto(s)
Adenoviridae/fisiología , Desmogleína 2/metabolismo , Virus Oncolíticos/fisiología , Internalización del Virus , Adenoviridae/genética , Apoptosis , Carcinoma Epitelial de Ovario/terapia , Línea Celular Tumoral , Ensayos Clínicos Fase I como Asunto , ADN Viral/sangre , Desmogleína 2/genética , Femenino , Humanos , Proteína Cofactora de Membrana/genética , Proteína Cofactora de Membrana/metabolismo , Viroterapia Oncolítica , Virus Oncolíticos/genética , Neoplasias Ováricas/terapia , Estudios Retrospectivos
3.
J Med Virol ; 91(9): 1702-1706, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31081549

RESUMEN

Melanoma, an immunogenic tumor, is the first indication where oncolytic viruses are now becoming part of clinical practice. ONCOS-102, a transgened adenovirus, has shown to act as a primer of relevant tumor targeting immune cells both in preclinical and clinical melanoma studies. Strategies to augment its effectiveness warrant investigation. Combination therapy of ONCOS-102 with the checkpoint inhibitor (CPI) pembrolizumab was evaluated in a quasi-human animal model, the humanized NOG mouse model. A dosing schedule of the combination, beginning the CPI concurrently with the oncolytic viral therapy and continuing the CPI treatment, appeared to induce an abscopal effect in untreated tumor lesions. Concurrent combination therapy with checkpoint inhibitors may improve the induction of antitumor immune responses of ONCOS-102.


Asunto(s)
Antineoplásicos Inmunológicos/farmacología , Melanoma/terapia , Viroterapia Oncolítica , Virus Oncolíticos , Animales , Línea Celular Tumoral , Terapia Combinada , Modelos Animales de Enfermedad , Vectores Genéticos/administración & dosificación , Vectores Genéticos/genética , Humanos , Melanoma/patología , Ratones , Ratones Transgénicos , Virus Oncolíticos/genética , Resultado del Tratamiento
4.
Int J Mol Sci ; 20(3)2019 Jan 31.
Artículo en Inglés | MEDLINE | ID: mdl-30709038

RESUMEN

Oncolytic adenoviruses can trigger lysis of tumor cells, induce an antitumor immune response, bypass classical chemotherapeutic resistance strategies of tumors, and provide opportunities for combination strategies. A major challenge is the development of scalable production methods for viral seed stocks and sufficient quantities of clinical grade viruses. Because of promising clinical signals in a compassionate use program (Advanced Therapy Access Program) which supported further development, we chose the oncolytic adenovirus ONCOS-401 as a testbed for a new approach to scale up. We found that the best viral production conditions in both T-175 flasks and HYPERFlasks included A549 cells grown to 220,000 cells/cm² (80% confluency), with ONCOS-401 infection at 30 multiplicity of infection (MOI), and an incubation period of 66 h. The Lysis A harvesting method with benzonase provided the highest viral yield from both T-175 and HYPERFlasks (10,887 ± 100 and 14,559 ± 802 infectious viral particles/cell, respectively). T-175 flasks and HYPERFlasks produced up to 2.1 × 108 ± 0.2 and 1.75 × 108 ± 0.08 infectious particles of ONCOS-401 per cm² of surface area, respectively. Our findings suggest a suitable stepwise process that can be applied to optimizing the initial production of other oncolytic viruses.


Asunto(s)
Adenoviridae/crecimiento & desarrollo , Virus Oncolíticos/crecimiento & desarrollo , Cultivo de Virus/instrumentación , Células A549 , Animales , Técnicas de Cultivo Celular por Lotes/instrumentación , Humanos , Carga Viral , Cultivo de Virus/métodos , Replicación Viral
5.
J Med Virol ; 90(10): 1669-1673, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-29797583

RESUMEN

Oncolytic adenoviral immunotherapy activates the innate immune system with subsequent induction of adaptive tumor-specific immune responses to fight cancer. Hence, oncolytic viruses do not only eradicate cancer cells by direct lysis, but also generate antitumor immune response, allowing for long-lasting cancer control and tumor reduction. Their therapeutic effect can be further enhanced by arming the oncolytic adenovirus with costimulatory transgenes and/or coadministration with other antitumor therapies. ONCOS-102 has already been found to be well tolerated and efficacious against some types of treatment-refractory tumors, including mesothelin-positive ovarian cancer (NCT01598129). It induced local and systemic CD8+ T-cell immunity and upregulated programmed death ligand 1. These results strongly advocate the use of ONCOS-102 in combination with other therapeutic strategies in advanced and refractory tumors, especially those expressing the mesothelin antigen. The in vivo work presented herein describes the ability of the oncolytic adenovirus ONCOS-102 to induce mesothelin-specific T-cells after the administration of the virus in bagg albino (BALB/c) mice with mesothelin-positive tumors. We also demonstrate the effectiveness of the interferon-γ the enzyme-linked immunospot (ELISPOT) assay to detect the induction of T-cells recognizing mesothelin, hexon, and E1A antigens in ONCOS-102-treated mesothelioma-bearing BALB/c mice. Thus, the ELISPOT assay could be useful to monitor the progress of therapy with ONCOS-102.


Asunto(s)
Adenoviridae/crecimiento & desarrollo , Anticuerpos Antineoplásicos/sangre , Mesotelioma/terapia , Viroterapia Oncolítica , Virus Oncolíticos/crecimiento & desarrollo , Animales , Modelos Animales de Enfermedad , Ensayo de Immunospot Ligado a Enzimas , Humanos , Mesotelina , Ratones Endogámicos BALB C , Linfocitos T/inmunología , Resultado del Tratamiento
6.
Mol Ther Oncolytics ; 28: 158-170, 2023 Mar 16.
Artículo en Inglés | MEDLINE | ID: mdl-36816748

RESUMEN

Immunotherapy of metastatic melanoma (MM) has vastly improved the longevity of only a minority of patients. To broaden the repertoire of agents against MM, we investigated the effectiveness of locally interrupting tumor blood endothelial cell proliferation and angiogenesis, arginine deprivation, or both on the growth of melanoma by constructing and characterizing the effectiveness of four oncolytic adenoviruses. ONCOS-207 (which expressed tissue inhibitor of metalloprotease type 2 [TIMP2]), ONCOS-209 (which expressed peptidyl arginine deiminase [PADI1]), and ONCOS-210 and ONCOS-212 (which expressed both TIMP2 and PADI1) exhibited oncolytic activity against four melanoma cell lines in vitro. ONCOS-212 treatments significantly inhibited tumor growth in an A2058 tumor model in nude mice compared with vehicle control. The inhibitory effects of the two transgenes of ONCOS-212 on tumor growth appeared to be synergistic. These viruses also significantly inhibited tumor growth in a humanized NOG model of melanoma (A2058 xenograft). All viruses significantly increased the percentage of activated CD8+ T cells in the tumor-infiltrating lymphocytes. The abscopal effect of ONCOS-212 treatments in the A2058 tumor challenge model in hNOG mice supports the hypothesis that the human immune response contributes to the anti-tumor activity of ONCOS-212. These results support the further development of ONCOS-212 for cancer treatment.

7.
Scand J Clin Lab Invest ; 69(2): 251-64, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-18951241

RESUMEN

OBJECTIVE: To investigate the early apoptosis that may be detected by Annexin V binding to phosphatidylserine and propidium iodide (PI) exclusion in human monocytes. When studying monocytes in culture, less than 40 % of these cells survive after 7 days. MATERIAL AND METHODS: In the first 4 h, 24 % of monocytes in culture develop into Annexin V(+)/PI(-) cells. Human monocytes were investigated at 0 h and sorted into Annexin V(+) and Annexin V(-) by FACS after 4 h. Gene expression was examined by microarray analyses. RESULTS: At 4 h, Annexin V(+) monocytes versus Annexin V(-) cells showed 1220 differentially expressed genes. Ingenuity Pathway Analysis disclosed 153 genes related to cell death. Among these were caspase activators, caspase 6, Apaf 2 and FAS, as well as the autophagy gene ATG5. In addition, examination of the most up-regulated or down-regulated genes among the 1220 revealed genes involved in other biological processes, as well as genes not yet annotated. These included the non-annotated genes LOC28480 (fold change: 82) and 225767-at (fold change: 68) and the transcription factor SOX 4 (fold change: 24). conclusions: We suggest that apoptosis in cultured monocytes, as evidenced by Annexin V(+), operates through genes well known in apoptosis, but that the process also involves additional genes not commonly associated with apoptosis.


Asunto(s)
Anexina A5/metabolismo , Perfilación de la Expresión Génica , Monocitos/metabolismo , Células Cultivadas , Citometría de Flujo , Humanos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
8.
Cancer Gene Ther ; 26(1-2): 26-31, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30057416

RESUMEN

Adaptive immunity involves activation of T cells via antigen presentation by antigen presenting cells (APCs) along with the action of co-stimulatory molecules and pattern recognition receptors. Cluster of differentiation 40 (CD40) is one such costimulatory molecule that is expressed on APCs that binds to CD40 ligand (CD40L) on T helper cells and activates a signaling cascade, subsequently resulting in a wide range of immune and inflammatory responses. Considering its important role in regulation of immune response, CD40/40 L has been used for developing antitumor vaccines. In this study, we developed methods for evaluating and quantifying the activity of CD40L expressed from an adenovirus vector ONCOS-401. Our results show that the ONCOS-401 vector produces functional CD40L, which can bind and activate a NF-κB-dependent signaling cascade, leading to secreted embryonic alkaline phosphatase reporter production in HEK293-BLUE cells. In addition, quantification of CD40L production using enzyme-linked immunosorbent assay and HEK-293 BLUE reporter cells showed reproducibly higher recovery of CD40L from ONCOS-401 than from the negative control vector or uninfected cells with consistent inter and intra-assay precision. Thus, a rapid and easy method for quantifying and assessing CD40L production and activity from adenovirus vectors would support the assessment of efficacy of the vector for gene therapy - this was the objective of our study.


Asunto(s)
Adenoviridae/genética , Ligando de CD40/genética , Vectores Genéticos , Fosfatasa Alcalina/metabolismo , Ligando de CD40/análisis , Células HEK293 , Humanos , FN-kappa B/metabolismo , Transducción de Señal
9.
Oncoimmunology ; 8(2): e1532763, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30713786

RESUMEN

Malignant melanoma is an aggressive type of skin cancer whose incidence is increasing globally. Although surgery is effective in early stage melanoma, patients with advanced melanoma only have a 20% 5-year survival rate. Hence, combinations of existing and new immunotherapy technologies and immunotherapeutic agents are being evaluated. ONCOS-102 is an oncolytic adenovirus armed with human GM-CSF and an Ad5/3 chimeric capsid. It has shown to be well tolerated in phase I study (NCT01598129) wherein it induced antitumor immunity, infiltration of CD8 + T cells to tumors, and up-regulation of PD-L1. We propose that ONCOS-102 could serve as an immunosensitizer in combination therapies with checkpoint inhibitors. In this preclinical study, we investigated the cytotoxicity of ONCOS-102 and pembrolizumab, an anti-PD-1 antibody, in four human melanoma cell lines, A375, A2058, SK-Mel-2 and SK-Mel-28. Humanized mice engrafted with A2058 melanoma cells showed significant tumor volume reduction after ONCOS-102 treatment. Combination of pembrolizumab with ONCOS-102 reduced tumor volume to an even greater extent, while pembrolizumab (200 µg, or 400 µg) did not show any therapeutic benefit by itself. Body weight loss, and metastasis were not significantly affected by any treatment. These data support the scientific rationale for the ongoing clinical study of combination therapy of ONCOS-102 and pembrolizumab for the treatment of melanoma (NCT03003676).

10.
Infect Immun ; 76(6): 2685-95, 2008 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-18362127

RESUMEN

Lipopolysaccharide (LPS) in the outer membrane of Neisseria meningitidis plays a dominant role as an inflammation-inducing molecule in meningococcal disease. We have used microarray analysis to study the global gene expression after exposure of human monocytes for 3 h to wild-type N. meningitidis (10(6)), LPS-deficient N. meningitidis (10(6) and 10(8)), and purified N. meningitidis LPS (1 ng [33 endotoxin units]/ml) to identify LPS-inducible genes. Wild-type N. meningitidis (10(6)) induced 4,689 differentially expressed genes, compared with 72 differentially expressed genes induced by 10(6) LPS-deficient N. meningitidis organisms. However, 10(8) LPS-deficient N. meningitidis organisms induced 3,905 genes, indicating a dose-response behavior of non-LPS cell wall molecules. A comparison of the gene expression patterns from 10(6) wild-type N. meningitidis and 10(8) LPS-deficient N. meningitidis organisms showed that 2,401 genes in human monocytes were not strictly LPS dependent. A list of "particularly LPS-sensitive" genes (2,288), differentially induced by 10(6) wild-type N. meningitidis but not by 10(8) LPS-deficient N. meningitidis organisms, showed an early expression of beta interferon (IFN-beta), most likely through the Toll-like receptor-MyD88-independent pathway. Subsequently, IFN-beta may activate the type I IFN signaling pathway, and an unknown number of IFN-beta-inducible genes, such as those for CXCL9, CXCL10, CXCL11, IFIT1, IFIT2, IFIT3, and IFIT5, are transcribed. Supporting this, human monocytes secreted significantly higher levels of CXCL10 and CXCL11 when stimulated by 10(6) wild-type N. meningitidis organisms than when stimulated by 10(8) LPS-deficient N. meningitidis organisms. Plasma CXCL10, but not CXCL11, was positively correlated (r = 0.67; P < 0.01) to LPS in patients (n = 24) with systemic meningococcal disease. Thus, new circulating biomarkers in meningococcal disease may be suggested through LPS-induced gene expression changes in human monocytes.


Asunto(s)
Perfilación de la Expresión Génica , Lipopolisacáridos/metabolismo , Lipopolisacáridos/farmacología , Monocitos/efectos de los fármacos , Monocitos/metabolismo , Neisseria meningitidis/genética , Quimiocina CXCL10/sangre , Quimiocina CXCL11/sangre , Regulación hacia Abajo , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Quinasas Janus/metabolismo , Infecciones Meningocócicas/sangre , Mutación , Neisseria meningitidis/fisiología , Factores de Transcripción STAT/metabolismo , Transducción de Señal/fisiología , Regulación hacia Arriba
11.
J Endotoxin Res ; 9(6): 349-60, 2003.
Artículo en Inglés | MEDLINE | ID: mdl-14733721

RESUMEN

Regulation of chemokine production and the expression of chemokine receptors play an important role during inflammation and infectious diseases. The present study was designed to study the effects of five different bacterial cell wall components (PAMPs) on the production of MCP-1 and MIP-1alpha and the expression of CCR2 by highly purified human blood monocytes. All five PAMPs induced high expression of mRNA and protein synthesis of both chemokines. Generally, MCP-1 mRNA and protein levels were higher than MIP-1alpha levels. Expression of MCP-1 and MIP-1alpha differed both at the mRNA and at the protein levels, MIP-1alpha always showing a more rapid initial increase, attaining lower protein levels than MCP-1. Antibodies against CD14 significantly inhibited the inducing effects of all the PAMPs used. Antibody against TLR2 inhibited the chemokine production induced by LTA and AraLAM by more than 36% (P < 0.05) while chemokine production induced by Escherichia coli-LPS, purified E. coli-LPS and Neisseria meningitidis-LPS was inhibited by more than 60% by antibody against TLR4 (P < 0.05). The inducing effects of all five PAMPs could be inhibited by rIL-4, rIL-10 and rIL-13. rIL-4 was the most effective. Generally, IC(50) of these anti-inflammatory cytokines were lower for the MIP-1alpha than for the MCP-1 production. The cell surface expression of CCR2 was significantly down-regulated by all five PAMPs in addition to a decrease in cytosolic free calcium and binding of rMCP-1. We conclude that MCP-1 and MIP-1alpha as well as the MCP-1 receptor CCR2 will be substantially regulated upon monocyte contact with various cell wall components (PAMPs) from Gram-negative and Gram-positive bacteria as well as from mycobacteria.


Asunto(s)
Quimiocina CCL2/metabolismo , Lipopolisacáridos/farmacología , Proteínas Inflamatorias de Macrófagos/metabolismo , Monocitos/metabolismo , Receptores de Quimiocina/metabolismo , Anticuerpos Monoclonales/metabolismo , Células Cultivadas , Quimiocina CCL2/genética , Quimiocina CCL3 , Quimiocina CCL4 , Citocinas/farmacología , Relación Dosis-Respuesta a Droga , Bacterias Gramnegativas/química , Bacterias Grampositivas/química , Humanos , Prueba de Limulus , Proteínas Inflamatorias de Macrófagos/genética , Monocitos/efectos de los fármacos , ARN Mensajero/genética , ARN Mensajero/metabolismo , Proteínas Recombinantes/metabolismo
12.
Am J Physiol Heart Circ Physiol ; 296(3): H787-95, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19136604

RESUMEN

Altered myocardial Ca(2+) and Na(+) handling in congestive heart failure (CHF) may be expected to decrease the tolerance to ischemia by augmenting reperfusion Ca(2+) overload. The aim of the present study was to investigate tolerance to hypoxia-reoxygenation by measuring enzyme release, cell death, ATP level, and cell Ca(2+) and Na(+) in cardiomyocytes from failing rat hearts. CHF was induced in Wistar rats by ligation of the left coronary artery during isoflurane anesthesia, after which cardiac failure developed within 6 wk. Isolated cardiomyocytes were cultured for 24 h and subsequently exposed to 4 h of hypoxia and 2 h of reoxygenation. Cell damage was measured as lactate dehydrogenase (LD) release, cell death as propidium iodide uptake, and ATP by firefly luciferase assay. Cell Ca(2+) and Na(+) were determined with radioactive isotopes, and free intracellular Ca(2+) concentration ([Ca(2+)](i)) with fluo-3 AM. CHF cells showed less increase in LD release and cell death after hypoxia-reoxygenation and had less relative reduction in ATP level after hypoxia than sham cells. CHF cells accumulated less Na(+) than sham cells during hypoxia (117 vs. 267 nmol/mg protein). CHF cells maintained much lower [Ca(2+)](i) than sham cells during hypoxia (423 vs. 1,766 arbitrary units at 4 h of hypoxia), and exchangeable Ca(2+) increased much less in CHF than in sham cells (1.4 vs. 6.7 nmol/mg protein) after 120 min of reoxygenation. Ranolazine, an inhibitor of late Na(+) current, significantly attenuated both the increase in exchangeable Ca(2+) and the increase in LD release in sham cells after reoxygenation. This supports the suggestion that differences in Na(+) accumulation during hypoxia cause the observed differences in Ca(2+) accumulation during reoxygenation. Tolerance to hypoxia and reoxygenation was surprisingly higher in CHF than in sham cardiomyocytes, probably explained by lower hypoxia-mediated Na(+) accumulation and subsequent lower Ca(2+) accumulation in CHF after reoxygenation.


Asunto(s)
Insuficiencia Cardíaca/metabolismo , Infarto del Miocardio/complicaciones , Daño por Reperfusión Miocárdica/prevención & control , Miocitos Cardíacos/metabolismo , Oxígeno/metabolismo , Acetanilidas/farmacología , Adenosina Trifosfato/metabolismo , Animales , Calcio/metabolismo , Muerte Celular , Hipoxia de la Célula , Células Cultivadas , Modelos Animales de Enfermedad , Insuficiencia Cardíaca/etiología , Insuficiencia Cardíaca/patología , L-Lactato Deshidrogenasa/metabolismo , Masculino , Infarto del Miocardio/metabolismo , Infarto del Miocardio/patología , Daño por Reperfusión Miocárdica/etiología , Daño por Reperfusión Miocárdica/metabolismo , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/enzimología , Miocitos Cardíacos/patología , Piperazinas/farmacología , Potasio/metabolismo , Ranolazina , Ratas , Ratas Wistar , Radioisótopos de Rubidio , Sodio/metabolismo , Bloqueadores de los Canales de Sodio/farmacología , Intercambiador de Sodio-Calcio/metabolismo , ATPasa Intercambiadora de Sodio-Potasio/metabolismo , Factores de Tiempo
13.
J Infect Dis ; 191(5): 768-75, 2005 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-15688294

RESUMEN

Chemokines are important in regulating leukocyte traffic during infection. We analyzed plasma chemokine levels of monocyte chemoattractant protein (MCP)-1, macrophage inflammatory protein (MIP)-1 alpha , interleukin (IL)-8, and RANTES in patients with meningococcal infection and correlated these to plasma lipopolysaccharide (LPS) levels, which are closely associated with clinical presentation. In patients with fulminant meningococcal septicemia, versus distinct meningitis or mild systemic meningococcal disease, MCP-1 (both P<.0001), MIP-1 alpha (both P<.0001), and IL-8 (P<.0001 and P=.011) were significantly higher and RANTES significantly lower (P=.007 and P=.021). MCP-1 (r=.88), MIP-1 alpha (r=.82), and IL-8 (r=.89) were positively correlated to plasma LPS levels, whereas RANTES was negatively correlated (r=-.49). In an ex vivo whole-blood model, heat-inactivated wild-type Neisseria meningitidis, purified meningococcal LPS, and (to a negligible extent) heat-inactivated LPS-deficient mutant N. meningitidis induced these chemokines. N. meningitidis LPS is the major cause of chemokine release in meningococcal disease.


Asunto(s)
Quimiocinas/sangre , Infecciones Meningocócicas/inmunología , Bacteriemia/inmunología , Bacteriemia/microbiología , Quimiocina CCL2/sangre , Quimiocina CCL4 , Quimiocina CCL5/sangre , Expresión Génica , Humanos , Interleucina-8/sangre , Lipopolisacáridos/inmunología , Proteínas Inflamatorias de Macrófagos/sangre , Neisseria meningitidis/inmunología , Factores de Tiempo
14.
Cytokine ; 32(6): 304-15, 2005 Dec 21.
Artículo en Inglés | MEDLINE | ID: mdl-16406558

RESUMEN

Recognition of conserved bacterial structures called pathogen-associated molecular patterns (PAMPs) by pattern recognition receptors (PRRs), may lead to induction of a variety of "early immediate genes" such as chemokines. In the current study, we have in an ex vivo whole blood model studied the induction of the chemokines MIP-1alpha, MCP-1 and IL-8 by various PAMPs. The rate of appearance of Escherichia coli-Lipopolysaccharide (LPS) induced chemokines differed. The production of MIP-1alpha and IL-8 was after 1 h of stimulation significantly higher when compared to unstimulated whole blood, whereas MCP-1 was not significantly elevated until after 3 h. At peak levels the MIP-1alpha concentration induced by E. coli-LPS was 3-5-fold higher than MCP-1 and IL-8. By specific cell depletion, we demonstrated that all three chemokines were mainly produced by monocytes. However, the mRNA results showed that IL-8 was induced in both monocytes and granulocytes. The production of all three chemokines, induced by the E. coli-LPS and Neisseria meningitidis-LPS, was significantly inhibited by antibodies against CD14 and TLR4, implying these receptors to be of importance for the effects of LPS in whole blood. The chemokine production induced by lipoteichoic acid (LTA) and non-mannose-capped lipoarabinomannan (AraLAM) was, however, less efficiently blocked by antibodies against CD14 and TLR2. E. coli-LPS and LTA induced a dose-dependent increase of CD14, TLR2 and TLR4 expression on monocytes in whole blood. These data show that PAMPs may induce chemokine production in whole blood and that antibodies against PRRs inhibit the production to different extent.


Asunto(s)
Quimiocinas/sangre , Polisacáridos Bacterianos/farmacología , Receptores de Reconocimiento de Patrones/sangre , Anticuerpos Monoclonales/farmacología , Antígenos Bacterianos/farmacología , Quimiocina CCL2/sangre , Quimiocina CCL2/genética , Quimiocina CCL3 , Quimiocina CCL4 , Quimiocinas/genética , Expresión Génica/efectos de los fármacos , Expresión Génica/genética , Granulocitos/efectos de los fármacos , Granulocitos/metabolismo , Heparina/farmacología , Humanos , Interleucina-8/sangre , Interleucina-8/genética , Procedimientos de Reducción del Leucocitos , Receptores de Lipopolisacáridos/inmunología , Receptores de Lipopolisacáridos/metabolismo , Lipopolisacáridos/farmacología , Proteínas Inflamatorias de Macrófagos/sangre , Proteínas Inflamatorias de Macrófagos/genética , Monocitos/efectos de los fármacos , Monocitos/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Ácidos Teicoicos/farmacología , Receptor Toll-Like 2/sangre , Receptor Toll-Like 2/inmunología , Receptor Toll-Like 4/sangre , Receptor Toll-Like 4/inmunología
15.
Infect Immun ; 72(6): 3344-9, 2004 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-15155639

RESUMEN

Fulminant meningococcal sepsis has been termed the prototypical lipopolysaccharide (LPS)-mediated gram-negative septic shock. Systemic inflammation by activated complement and cytokines is important in the pathogenesis of this disease. We investigated the involvement of meningococcal LPS in complement activation, complement-dependent inflammatory effects, and cytokine or chemokine production. Whole blood anticoagulated with lepirudin was stimulated with wild-type Neisseria meningitidis H44/76 (LPS+), LPS-deficient N. meningitidis H44/76lpxA (LPS-), or purified meningococcal LPS (NmLPS) at concentrations that were relevant to meningococcal sepsis. Complement activation products, chemokines, and cytokines were measured by enzyme-linked immunosorbent assays, and granulocyte CR3 (CD11b/CD18) upregulation and oxidative burst were measured by flow cytometry. The LPS+ and LPS- N. meningitidis strains both activated complement effectively and to comparable extents. Purified NmLPS, used at a concentration matched to the amount present in whole bacteria, did not induce any complement activation. Both CR3 upregulation and oxidative burst were also induced, independent of LPS. Interleukin-1beta (IL-1beta), tumor necrosis factor alpha, and macrophage inflammatory protein 1alpha production was predominantly dependent on LPS, in contrast to IL-8 production, which was also markedly induced by the LPS- meningococci. In this whole blood model of meningococcal sepsis, complement activation and the immediate complement-dependent inflammatory effects of CR3 upregulation and oxidative burst occurred independent of LPS.


Asunto(s)
Activación de Complemento , Inflamación/fisiopatología , Lipopolisacáridos/farmacología , Infecciones Meningocócicas/fisiopatología , Neisseria meningitidis/patogenicidad , Proteínas del Sistema Complemento/farmacología , Citocinas/biosíntesis , Granulocitos/metabolismo , Humanos , Antígeno de Macrófago-1/metabolismo , Modelos Biológicos , Estallido Respiratorio , Sepsis/fisiopatología , Regulación hacia Arriba
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA