Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 45
Filtrar
1.
Int J Pharm ; 586: 119498, 2020 Aug 30.
Artículo en Inglés | MEDLINE | ID: mdl-32505575

RESUMEN

Solid tumors are intrinsically resistant to immunotherapy because of the major challenges including the immunosuppression and poor penetration of drugs and lymphocytes into solid tumors due to the complicated tumor microenvironment (TME). Our previous study has created a novel superantigen mutant ST-4 to efficiently active the T lymphocytes and alleviate immune suppression. In the present study, to accumulate ST-4 into the TME, we constructed a recombinant protein, ST-4-iRGD, by fusing ST-4 to a tumor-homing peptide, iRGD. We hypothesized that ST-4-iRGD could internalize into the TME through iRGD-mediated tumor targeting and tumor tissue penetrating to activate the regional immunoreaction. The results of in vitro studies showed that ST-4-iRGD achieved improved tumor targeting and cytotoxicity in mouse B16F10 melanoma cells. The iRGD-mediated tumor tissue penetration was further confirmed by imaging and immunofluorescence studies in vivo, wherein higher distribution of ST-4-iRGD was observed in the mouse 4T1 breast tumor model. Moreover, ST-4-iRGD exhibited enhanced anti-solid tumor characteristics and induced improved lymphocyte infiltration in the B16F10 and 4T1 models. In conclusion, using iRGD to facilitate better dissemination of the therapeutic agent ST-4 throughout a solid tumor mass is feasible, and ST-4-iRGD may be a potential candidate for efficient cancer immunotherapy in the future.


Asunto(s)
Neoplasias de la Mama/terapia , Inmunoterapia/métodos , Melanoma Experimental/terapia , Oligopéptidos/administración & dosificación , Superantígenos/administración & dosificación , Animales , Neoplasias de la Mama/inmunología , Línea Celular Tumoral , Femenino , Melanoma Experimental/inmunología , Ratones , Ratones Endogámicos BALB C , Mutación , Superantígenos/genética , Superantígenos/metabolismo , Linfocitos T/inmunología , Microambiente Tumoral , Ensayos Antitumor por Modelo de Xenoinjerto
2.
FASEB J ; 33(6): 6919-6932, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30817226

RESUMEN

T cells expressing invariant γδ antigen receptors (γδ T cells) bridge innate and adaptive immunity and facilitate barrier responses to pathogens. Macrophage migration inhibitory factor (MIF) is an upstream mediator of host defense that up-regulates the expression of pattern recognition receptors and sustains inflammatory responses by inhibiting activation-induced apoptosis in monocytes and macrophages. Surprisingly, Mif-/- γδ T cells, when compared with wild type, were observed to produce >10-fold higher levels of the proinflammatory cytokine IL-17 after stimulation with gram-positive exotoxins. High-IL-17 expression was associated with the characteristic features of IL-17-producing γδ T (γδ17) cells, including expression of IL-23R, IL-1R1, and the transcription factors RORγt and Sox13. In the gram-positive model of shock mediated by toxic shock syndrome toxin (TSST-1), Mif-/- mice succumbed to death more quickly with increased pulmonary neutrophil accumulation and higher production of cytokines, including IL-1ß and IL-23. Mif-/- γδ T cells also produced high levels of IL-17 in response to Mycobacterium lipomannan, and depletion of γδ T cells improved survival from acutely lethal Mycobacterium infection or TSST-1 administration. These data indicate that MIF deficiency is associated with a compensatory amplification of γδ17 cell responses, with implications for innate immunity and IL-17-mediated pathology in situations such as gram-positive toxic shock or Mycobacterium infection.-Kim, H. K., Garcia, A. B., Siu, E., Tilstam, P., Das, R., Roberts, S., Leng, L., Bucala, R. Macrophage migration inhibitory factor regulates innate γδ T-cell responses via IL-17 expression.


Asunto(s)
Inmunidad Innata/inmunología , Inflamación/inmunología , Interleucina-17/metabolismo , Oxidorreductasas Intramoleculares/fisiología , Factores Inhibidores de la Migración de Macrófagos/fisiología , Receptores de Antígenos de Linfocitos T gamma-delta/inmunología , Células Th17/inmunología , Tuberculosis Pulmonar/inmunología , Animales , Toxinas Bacterianas/administración & dosificación , Enterotoxinas/administración & dosificación , Inflamación/metabolismo , Inflamación/patología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Mycobacterium bovis/inmunología , Receptores de Interleucina/metabolismo , Choque Séptico/inducido químicamente , Choque Séptico/inmunología , Choque Séptico/patología , Superantígenos/administración & dosificación , Tuberculosis Pulmonar/microbiología , Tuberculosis Pulmonar/patología
3.
Tumour Biol ; 37(4): 5305-16, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26561468

RESUMEN

Recent research has attempted to direct superantigens towards tumors by means of tumor-targeted superantigen (TTS) strategy. In this study, we explored the antitumor property of TTS by fusing the third loop of transforming growth factor α (TGFαL3) to staphylococcal enterotoxin type B (SEB) and investigated the possibility of the therapeutic application of TGFαL3-SEB as a novel antitumor candidate in mice bearing breast cancer. Treatment was performed through intratumoral and intravenous injection of TGFαL3-SEB. Tumor size/volume, long-term survival, and cytokine secretion were assessed. In addition, the toxicity of each treatment on liver and kidneys was examined. Our results indicated that the relative tumor volume significantly increased in the mice receiving intratumoral TGFaL3-SEB (p < 0.05). Surprisingly, 5 out of the 14 mice were cleared from the tumor thoroughly in 10-25 days after intratumoral administration of TGFaL3-SEB. Quantification of cytokines clearly showed that the mice receiving intratumoral SEB significantly secreted higher interferon γ (IFN-γ) and tumor necrosis factor α (TNF-α) compared with the other groups (p < 0.05). The antitumor effect was followed by inhibition of cell proliferation (Ki-67) and micro vascularization (CD31). The highest and lowest levels of tumor necrosis were observed in the intratumoral administration of TGFαL3-SEB (85 %) and PBS (14 %), respectively. Intratumoral injection of TGFαL3-SEB increased the lifespan of the mice so 37.5 % of them could survive for more than 6 months (p < 0.05). Overall, our findings indicated that intratumoral administration of TGFαL3-SEB effectively inhibited the growth of breast tumors through induction of necrosis and suppressing proliferation and angiogenesis without systemic toxicity.


Asunto(s)
Neoplasias de la Mama/terapia , Proliferación Celular/efectos de los fármacos , Enterotoxinas/administración & dosificación , Neovascularización Patológica/terapia , Factor de Necrosis Tumoral alfa/administración & dosificación , Animales , Neoplasias de la Mama/genética , Neoplasias de la Mama/inmunología , Enterotoxinas/genética , Enterotoxinas/inmunología , Femenino , Humanos , Inmunoterapia/métodos , Interferón gamma/metabolismo , Ratones , Neovascularización Patológica/inmunología , Proteínas de Fusión Oncogénica , Superantígenos/administración & dosificación , Superantígenos/inmunología , Factor de Crecimiento Transformador alfa/metabolismo , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/inmunología , Ensayos Antitumor por Modelo de Xenoinjerto
4.
Eur Rev Med Pharmacol Sci ; 19(13): 2400-5, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-26214775

RESUMEN

OBJECTIVE: To investigate the anti-glioma effects of staphylococcal enterotoxin C (SEC) combined with surgery, radiotherapy and chemotherapy, and the IL-2 of lymphocytes activated by SEC. PATIENTS AND METHODS: A total of 92 patients with gliomas which had been surgically removed, were randomly divided into a control, and treatment groups A (systemic application of SEC) and B (local application of lymphocytes activated by SEC). The treatment effects were observed on a CT scan of the head. The SEC's induction on the IL-2 secretion by lymphocytes was determined using immunohistochemistry and RT-PCR methods. RESULTS: Clinical materials showed that the effective rate (CR and PR) in the control group was 32.0%. In the treatment group A, the effective rate was 51.6% and 63.6% in the group B. The IL-2 of lymphocytes secreting activity activated by SEC began to secrete IL-2 on the 1st day and reached a peak during the 3rd day and the 5th day. CONCLUSIONS: As a superantigen, SEC combined with surgery, radiotherapy and chemotherapy can improve treatment. The lymphocytes activated by SEC can release IL-2.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/administración & dosificación , Neoplasias Encefálicas/terapia , Quimioradioterapia/métodos , Enterotoxinas/administración & dosificación , Glioma/terapia , Superantígenos/administración & dosificación , Adolescente , Adulto , Anciano , Neoplasias Encefálicas/diagnóstico , Terapia Combinada/métodos , Femenino , Estudios de Seguimiento , Glioma/diagnóstico , Humanos , Masculino , Persona de Mediana Edad , Resultado del Tratamiento , Adulto Joven
5.
Vaccine ; 30(34): 5099-109, 2012 Jul 20.
Artículo en Inglés | MEDLINE | ID: mdl-22691432

RESUMEN

Staphylococcus aureus causes significant illnesses throughout the world, including toxic shock syndrome (TSS), pneumonia, and infective endocarditis. Major contributors to S. aureus illnesses are secreted virulence factors it produces, including superantigens and cytolysins. This study investigates the use of superantigens and cytolysins as staphylococcal vaccine candidates. Importantly, 20% of humans and 50% of rabbits in our TSS model cannot generate antibody responses to native superantigens. We generated three TSST-1 mutants; G31S/S32P, H135A, and Q136A. All rabbits administered these TSST-1 toxoids generated strong antibody responses (titers>10,000) that neutralized native TSST-1 in TSS models, both in vitro and in vivo. These TSST-1 mutants lacked detectable residual toxicity. Additionally, the TSST-1 mutants exhibited intrinsic adjuvant activity, increasing antibody responses to a second staphylococcal antigen (ß-toxin). This effect may be due to TSST-1 mutants binding to the immune co-stimulatory molecule CD40. The superantigens TSST-1 and SEC and the cytolysin α-toxin are known to contribute to staphylococcal pneumonia. Immunization of rabbits against these secreted toxins provided complete protection from highly lethal challenge with a USA200 S. aureus strain producing all three exotoxins; USA200 strains are common causes of staphylococcal infections. The same three exotoxins plus the cytolysins ß-toxin and γ-toxin contribute to infective endocarditis and sepsis caused by USA200 strains. Immunization against these five exotoxins protected rabbits from infective endocarditis and lethal sepsis. These data suggest that immunization against toxoid proteins of S. aureus exotoxins protects from serious illnesses, and concurrently superantigen toxoid mutants provide endogenous adjuvant activity.


Asunto(s)
Toxinas Bacterianas/inmunología , Citotoxinas/inmunología , Proteínas Hemolisinas/inmunología , Conejos/inmunología , Infecciones Estafilocócicas/terapia , Staphylococcus aureus/inmunología , Superantígenos/inmunología , Adyuvantes Inmunológicos/administración & dosificación , Animales , Anticuerpos Antibacterianos/sangre , Anticuerpos Antibacterianos/inmunología , Formación de Anticuerpos , Proteínas Bacterianas/inmunología , Toxinas Bacterianas/administración & dosificación , Antígenos CD40/inmunología , Línea Celular , Citotoxinas/administración & dosificación , Endocarditis Bacteriana/inmunología , Endocarditis Bacteriana/microbiología , Endocarditis Bacteriana/terapia , Exotoxinas/inmunología , Femenino , Proteínas Hemolisinas/administración & dosificación , Humanos , Masculino , Pruebas de Neutralización , Neumonía Estafilocócica/inmunología , Neumonía Estafilocócica/microbiología , Neumonía Estafilocócica/terapia , Conejos/microbiología , Choque Séptico/inmunología , Choque Séptico/microbiología , Choque Séptico/terapia , Infecciones Estafilocócicas/inmunología , Infecciones Estafilocócicas/microbiología , Toxoide Estafilocócico/administración & dosificación , Toxoide Estafilocócico/inmunología , Vacunas Estafilocócicas/inmunología , Staphylococcus aureus/patogenicidad , Superantígenos/administración & dosificación , Vacunación
6.
Mol Vis ; 18: 974-82, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22550390

RESUMEN

PURPOSE: We assessed the combined use of Staphylococcal Enterotoxin B (SEB) superantigen pre-treatment along with allogeneic bone marrow transplant (BMT) to induce immune suppression condition and inhibit corneal keratoplasty rejection in mice. METHODS: BALB/C (H-2d) mice were both BMT and corneal allografts donors and C57BL/6(H-2b) mice were recipients. Prior to BMT, recipients received single injections of either SEB, cyclophosphamide (CYP), or normal saline (NS). Allogenic corneal penetrating keratoplasty was performed 7 days after BMT. Bone marrow chimerisms in recipients (donor major histocompatibility complex-II H2-d) were determined on Days 14, 28, and 56 post-BMT. Recipient immune response was assessed by mixed lymphocyte reactions (MLR) using splenocytes from C57BL/6 mice as responders in co-culture with stimulator cells from C57BL/6 (isogeneic), BALB/C (allogeneic), or CBA/1(third party) mice. Cluster of differentiation 4 receptors positive (CD4+) and CD8+T cells in recipient mice were evaluated. Corneal graft survival was assessed using Kaplan-Meier survival curves. RESULTS: SEB pre-treatment induced higher levels of hematopoietic chimerism on Days 14, 28 and 56 post-BMT than did CYP or NS pre-treatment. Mean corneal allograft survival was significantly prolonged with group SEB-BMT (20.3±7.6 days) compared to group CYP-BMT (13.0±4.0 days) and NS-BMT (9.0±2.2 days). SEB-BMT mice splenocytes had diminished MLR responses compared to CYP-BMT or NS-BMT mice. CD4+ and CD8+ T cells in peripheral blood and spleens were significantly reduced in group SEB-BMT mice. CONCLUSIONS: BMT after SEB pre-treatment could promote mixed chimerism, which inhibited allogeneic cornea transplant rejection. This should possibly relate to CD4+ and CD8+ T cell deletion and acquiring donor-specific immunosuppression.


Asunto(s)
Trasplante de Médula Ósea , Córnea/inmunología , Enterotoxinas/administración & dosificación , Supervivencia de Injerto/efectos de los fármacos , Superantígenos/administración & dosificación , Acondicionamiento Pretrasplante/métodos , Animales , Técnicas de Cocultivo , Córnea/cirugía , Trasplante de Córnea , Ciclofosfamida/administración & dosificación , Femenino , Supervivencia de Injerto/inmunología , Tolerancia Inmunológica/efectos de los fármacos , Terapia de Inmunosupresión/métodos , Estimación de Kaplan-Meier , Prueba de Cultivo Mixto de Linfocitos , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Bazo/citología , Bazo/efectos de los fármacos , Bazo/inmunología , Linfocitos T/efectos de los fármacos , Linfocitos T/inmunología , Quimera por Trasplante/inmunología , Trasplante Homólogo
7.
PLoS Biol ; 9(9): e1001149, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21931534

RESUMEN

Bacterial superantigens, a diverse family of toxins, induce an inflammatory cytokine storm that can lead to lethal shock. CD28 is a homodimer expressed on T cells that functions as the principal costimulatory ligand in the immune response through an interaction with its B7 coligands, yet we show here that to elicit inflammatory cytokine gene expression and toxicity, superantigens must bind directly into the dimer interface of CD28. Preventing access of the superantigen to CD28 suffices to block its lethality. Mice were protected from lethal superantigen challenge by short peptide mimetics of the CD28 dimer interface and by peptides selected to compete with the superantigen for its binding site in CD28. Superantigens use a conserved ß-strand/hinge/α-helix domain of hitherto unknown function to engage CD28. Mutation of this superantigen domain abolished inflammatory cytokine gene induction and lethality. Structural analysis showed that when a superantigen binds to the T cell receptor on the T cell and major histocompatibility class II molecule on the antigen-presenting cell, CD28 can be accommodated readily as third superantigen receptor in the quaternary complex, with the CD28 dimer interface oriented towards the ß-strand/hinge/α-helix domain in the superantigen. Our findings identify the CD28 homodimer interface as a critical receptor target for superantigens. The novel role of CD28 as receptor for a class of microbial pathogens, the superantigen toxins, broadens the scope of pathogen recognition mechanisms.


Asunto(s)
Antígenos CD28/inmunología , Citocinas/genética , Choque Séptico/inmunología , Superantígenos/inmunología , Secuencia de Aminoácidos , Animales , Toxinas Bacterianas/inmunología , Antígenos CD28/genética , Línea Celular Tumoral , Citocinas/inmunología , Enterotoxinas/inmunología , Mapeo Epitopo , Escherichia coli/genética , Escherichia coli/metabolismo , Femenino , Regulación de la Expresión Génica , Vectores Genéticos , Humanos , Inmunidad Celular , Leucocitos Mononucleares/citología , Leucocitos Mononucleares/inmunología , Ratones , Ratones Endogámicos BALB C , Datos de Secuencia Molecular , Unión Proteica , Receptores de Antígenos de Linfocitos T/inmunología , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/inmunología , Choque Séptico/genética , Staphylococcus aureus/genética , Staphylococcus aureus/inmunología , Superantígenos/administración & dosificación , Resonancia por Plasmón de Superficie
8.
Blood ; 117(18): 4778-86, 2011 May 05.
Artículo en Inglés | MEDLINE | ID: mdl-21406724

RESUMEN

An important role for natural killer (NK) cells in the regulation of T-cell responses is emerging, although the receptor pairs regulating the NK-T-cell interaction have still not been identified. We found that superantigen-stimulated T cells express Nectin-2 (CD112) and poliovirus receptor (PVR; CD155), the ligands of the activating NK receptor DNAX accessory molecule-1 (DNAM-1; CD226). Interestingly, only PVR was present at the T cell surface, particularly on cells in the S and G(2)/M phases of the cell cycle. The up-regulation of PVR expression involves DNA-damage response (DDR)-dependent pathways, because we found that pharmacologic inhibition of ATM and ATR kinases reduced PVR expression and that PVR was almost exclusively induced on cells expressing the DDR marker γH2AX. Oxidative stress contributed to DDR activation, and our results showed impaired PVR levels in the presence of the reactive oxygen species (ROS) scavenger N-acetyl-cysteine (NAC), being monocytes the main ROS source needed for optimal PVR expression on activated T cells. Interestingly, in accordance with ligand expression, NK cells lysed allogeneic proliferating more efficiently than nonproliferating T lymphocytes, with a mechanism requiring the cooperation between DNAM-1 and NKG2D. These results could contribute to unraveling the role of NK cells in the down-regulation of T-cell responses in physiologic and pathologic processes such as autoimmunity or GVHD.


Asunto(s)
Antígenos de Diferenciación de Linfocitos T/metabolismo , Daño del ADN , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/metabolismo , Linfocitos T/inmunología , Linfocitos T/metabolismo , Proteínas de la Ataxia Telangiectasia Mutada , Secuencia de Bases , Moléculas de Adhesión Celular/biosíntesis , Moléculas de Adhesión Celular/genética , Proteínas de Ciclo Celular/metabolismo , Proliferación Celular , Citotoxicidad Inmunológica , Cartilla de ADN/genética , Proteínas de Unión al ADN/metabolismo , Humanos , Técnicas In Vitro , Receptores de Lipopolisacáridos/metabolismo , Activación de Linfocitos , Cooperación Linfocítica , Subfamilia K de Receptores Similares a Lectina de Células NK/metabolismo , Nectinas , Estrés Oxidativo , Proteínas Serina-Treonina Quinasas/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Receptores Virales/biosíntesis , Receptores Virales/genética , Superantígenos/administración & dosificación , Linfocitos T/citología , Proteínas Supresoras de Tumor/metabolismo , Regulación hacia Arriba
9.
J Immunother ; 33(5): 492-9, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20463598

RESUMEN

Antibody-targeted superantigens have a potential to become useful drugs for tumor therapy. However, clinical practice has identified several issues that need to be addressed to optimize such molecules. On the basis of the experience from superantigen products in clinical trials, a novel tumor-targeted superantigen, naptumomab estafenatox (5T4FabV18-SEA/E-120 or ABR-217620) has been designed. Critical properties, such as tumor reactivity, therapeutic window, and seroreactivity were all improved. The engineered 5T4Fab moiety recognizes the 5T4 antigen expressed on a large number of solid tumor forms with an affinity in the order of 1 nM. The fusion protein induces T-cell mediated killing of tumor cells at concentrations around 10 pM. Compared with a construct with a wild-type superantigen, it is more potent in mediating killing of tumor cells but a 10,000-fold less active in mediating killing of MHC class II positive cells. The target epitopes for naturally occurring antibodies toward bacterial superantigens are reduced. Only large excesses of human anti-SEA antibodies neutralize the antitumor effects of the antibody-targeted superantigen. Naptumomab estafenatox induces dramatic reduction of established human tumors in Severe Combined Immunodeficient mice grafted with human lymphocytes. Thus, naptumomab estafenatox is a novel optimized tumor-targeted superantigen currently investigated in clinical trials.


Asunto(s)
Anticuerpos Monoclonales/administración & dosificación , Carcinoma de Células Renales/inmunología , Enterotoxinas/administración & dosificación , Inmunotoxinas/uso terapéutico , Proteínas Recombinantes de Fusión/administración & dosificación , Linfocitos T/metabolismo , Animales , Anticuerpos Monoclonales/genética , Anticuerpos Monoclonales/metabolismo , Antígenos de Neoplasias/genética , Antígenos de Neoplasias/inmunología , Antígenos de Neoplasias/metabolismo , Carcinoma de Células Renales/tratamiento farmacológico , Carcinoma de Células Renales/patología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Citotoxicidad Inmunológica/efectos de los fármacos , Enterotoxinas/genética , Enterotoxinas/metabolismo , Humanos , Inmunoconjugados , Macaca fascicularis , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/inmunología , Glicoproteínas de Membrana/metabolismo , Ratones , Ratones SCID , Trasplante de Neoplasias , Mutación Puntual/genética , Ingeniería de Proteínas , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Superantígenos/administración & dosificación , Superantígenos/genética , Superantígenos/metabolismo , Linfocitos T/efectos de los fármacos , Linfocitos T/inmunología , Linfocitos T/patología
10.
J Immunol ; 182(3): 1260-9, 2009 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-19155471

RESUMEN

An efficient pathway of cross-presentation common to a range of dendritic cell (DC) populations was identified by targeting Ag to MHC class II molecules. This finding was achieved by conjugating Ag to M1, which is a modified version of the superantigen streptococcal mitogenic exotoxin Z-2 that binds to MHC class II molecules but cannot directly stimulate T cells. M1 conjugates were efficiently presented to CD4(+) and CD8(+) T cells by bone marrow-derived DC and Langerhans cells in vitro. Whereas nonconjugated Ag was preferentially cross-presented by splenic CD8alpha(+) DC in vivo, M1-conjugated Ag was cross-presented by all dendritic subtypes assessed. Potent effector T cell responses with antitumor activity were elicited when M1 conjugates were injected together with an adjuvant. This method of Ag delivery has significant potential in therapeutic applications.


Asunto(s)
Toxinas Bacterianas/administración & dosificación , Toxinas Bacterianas/inmunología , Reactividad Cruzada/inmunología , Sistemas de Liberación de Medicamentos/métodos , Exotoxinas/administración & dosificación , Exotoxinas/inmunología , Antígenos de Histocompatibilidad Clase II/metabolismo , Melanoma Experimental/inmunología , Melanoma Experimental/terapia , Animales , Presentación de Antígeno/inmunología , Células Presentadoras de Antígenos/inmunología , Células Presentadoras de Antígenos/metabolismo , Toxinas Bacterianas/metabolismo , Vacunas contra el Cáncer/administración & dosificación , Vacunas contra el Cáncer/inmunología , Vacunas contra el Cáncer/metabolismo , Línea Celular Tumoral , Células Cultivadas , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Exotoxinas/metabolismo , Ligandos , Melanoma Experimental/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Transducción de Señal/inmunología , Superantígenos/administración & dosificación , Superantígenos/inmunología , Superantígenos/metabolismo
11.
Int Immunopharmacol ; 8(3): 442-52, 2008 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18279798

RESUMEN

In this study we explored the possibility of improving the anti-tumor potency of tumor-targeted superantigens (TTS) by combination treatment with interferon-alpha (IFN-alpha). TTS utilizes the powerful T cell activating property of the superantigen staphylococcal enterotoxin A (SEA) in fusion with an anti-tumor Fab-fragment to target this T cell activity against tumor cells. TTS fusion proteins have shown anti-tumor efficacy in a number of experimental tumor models including the B16 mouse melanoma transfected with a human tumor-associated antigen recognized by the C215 monoclonal antibody. IFN-alpha is approved for the treatment of solid tumors such as renal cell carcinoma and malignant melanoma and exerts immunomodulatory effects, which make it an appropriate candidate to combine with immunotherapy against cancer. Here we report that daily administration of IFN-alpha (20 000 U i.p.) enhances and sustains CD8+ T cell activation induced by the TTS C215Fab-SEA (10 microg i.v.) in C57Bl/6 mice, as reflected by increased and prolonged cell-mediated cytotoxicity against tumor cells ex vivo as well as by augmented serum IFN-gamma levels. C215Fab-SEA synergized with IFN-alpha in reducing the number of lung tumors in B16-C215 melanoma bearing mice as compared to mono therapy. In a long term tumor survival experiment, the prolonged median survival time of the combination treatment was 3.5 and 7.7 times the prolonged median survival times of C215Fab-SEA and IFN-alpha monotherapies, respectively. Hence, the combination treatment provoked synergistic anti-tumor effects as measured by the number of lung tumors and markedly prolonged survival. The enhanced therapeutic efficacy correlated with a striking and sustained increase of CD8- and perforin-expressing tumor-infiltrating cells. These results suggest significant potential of combining TTS with IFN-alpha for human cancer therapy.


Asunto(s)
Enterotoxinas/administración & dosificación , Fragmentos Fab de Inmunoglobulinas/administración & dosificación , Interferón-alfa/administración & dosificación , Melanoma Experimental/terapia , Superantígenos/administración & dosificación , Animales , Línea Celular Tumoral , Citotoxicidad Inmunológica , Femenino , Interferón gamma/biosíntesis , Linfocitos Infiltrantes de Tumor/inmunología , Melanoma Experimental/inmunología , Ratones , Ratones Endogámicos C57BL , Linfocitos T Citotóxicos/inmunología
12.
Res Vet Sci ; 83(2): 182-7, 2007 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17412377

RESUMEN

Evaluation of drug efficacy for human diseases is routinely performed in animal models for efficiency and in accordance with FDA regulations. Rhesus macaques have been used as models for various lethal diseases and correlates of immunity, as nonhuman primates (NHP) closely resemble humans. We examined the ex vivo cytokine response of superantigen-stimulated whole-blood cells as a first step to therapeutic efficacy testing for bacterial superantigen-induced shock in NHP after oral dosing of pentoxifylline. Doses of 120mg/kg of pentoxifylline effectively attenuated staphylococcal enterotoxin B-induced tumor necrosis factor alpha (TNFalpha), gamma interferon (IFNgamma) and interleukin 2 (IL-2) in ex vivo culture of NHP whole-blood cells by 88%, 81%, and 76%, respectively, whereas lower doses of 48 or 72mg/kg had no inhibitory effect. Thus cytokine release of stimulated peripheral blood cells provides a convenient biological measurement of the anti-inflammatory potency of pentoxifylline and has the advantage of assessing functional responses to a specific biotoxin of interest.


Asunto(s)
Células Sanguíneas/efectos de los fármacos , Células Sanguíneas/metabolismo , Citocinas/metabolismo , Macaca mulatta , Pentoxifilina/farmacología , Superantígenos/farmacología , Administración Oral , Animales , Células Sanguíneas/inmunología , Células Cultivadas , Relación Dosis-Respuesta a Droga , Enterotoxinas/toxicidad , Superantígenos/administración & dosificación , Factores de Tiempo , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores
13.
J Korean Med Sci ; 21(2): 315-23, 2006 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-16614521

RESUMEN

Staphylococcus aureus may perform an crucial function in atopic dermatitis (AD), via the secretion of superantigens, including staphylococcal enterotoxins (SE) A or B, and toxic shock syndrome toxin-1 (TSST-1). Dysregulated cytokine production by keratinocytes (KCs) upon exposure to staphylococcal superantigens (SsAgs) may be principally involved in the pathophysiology of AD. We hypothesized that lesional KCs from AD may react differently to SsAgs compared to nonlesional skin or normal skin from nonatopics. We conducted a comparison of HLA-DR or CD1a expression in lesional skin as opposed to that in nonlesional or normal skin by immunohistochemistry (IHC). We also compared, using ELISA, the levels of IL-1alpha, IL-1beta, and TNF-alpha secreted by cultured KCs from lesional, nonlesional, and normal skin, after the addition of SEA, SEB and TSST-1. IHC revealed that both HLA-DR and CD1a expression increased significantly in the epidermis of lesional skin versus nonlesional or normal skin in quite a similar manner. IL-1alpha, IL-1beta, and TNF-alpha secretion was also significantly elevated in the cultured KCs from lesional skin after the addition of SsAgs. Our results indicated that KCs from lesional skin appear to react differently to SsAgs and increased proinflammatory cytokine production in response to SsAgs may contribute to the pathogenesis of AD.


Asunto(s)
Dermatitis Atópica/microbiología , Queratinocitos/microbiología , Staphylococcus aureus/inmunología , Superantígenos , Adulto , Antígenos CD1/metabolismo , Toxinas Bacterianas/administración & dosificación , Toxinas Bacterianas/inmunología , Secuencia de Bases , Estudios de Casos y Controles , ADN Complementario/genética , Dermatitis Atópica/etiología , Dermatitis Atópica/inmunología , Enterotoxinas/administración & dosificación , Enterotoxinas/inmunología , Antígenos HLA-DR/metabolismo , Humanos , Técnicas In Vitro , Mediadores de Inflamación/metabolismo , Interleucina-1/biosíntesis , Interleucina-1/genética , Queratinocitos/inmunología , Masculino , Staphylococcus aureus/patogenicidad , Superantígenos/administración & dosificación , Superantígenos/inmunología , Factor de Necrosis Tumoral alfa/biosíntesis , Factor de Necrosis Tumoral alfa/genética
14.
Infect Immun ; 74(2): 1196-203, 2006 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-16428769

RESUMEN

Staphylococcal protein A (SpA) is representative of a new class of antigens, the B-cell superantigens (SAgs). These antigens bind to the Fab regions of immunoglobulin molecules outside their complementarity-determining regions. SpA, the best-studied B-cell SAg, reacts with the Fabs of most VH3+ immunoglobulins, which are expressed on 30 to 60% of human peripheral B cells. Therefore, B-cell SAgs like SpA have great potential to elicit inflammatory responses in vivo. We previously reported that the interaction of SpA with VH3+ immunoglobulin molecules leads to activation of the complement cascade and produces a histologic pattern of inflammation in the skin of a rabbit indicative of immune complex injury. To elucidate the cellular and molecular events contributing to this type of unconventional immune complex-mediated inflammation, we established a mouse peritoneal Arthus reaction model. Mice treated intravenously with human polyclonal immunoglobulin G (IgG), followed by intraperitoneal injection of SpA, showed neutrophil influx into the peritoneal cavity with peak numbers appearing at 8 h. This inflammatory reaction was dependent on the interaction of SpA with VH3+ IgG. Mast cells, FcgammaRIII, complement components, and tumor necrosis factor alpha play obligatory roles, and the reaction is associated with the local release of the CXC chemokines macrophage inflammatory protein 2 and KC. The data provide further compelling evidence for the induction of immune complex-mediated injury by a B-cell SAg and highlight important factors contributing to the pathogenesis of this novel type of inflammatory reaction.


Asunto(s)
Reacción de Arthus/inmunología , Reacción de Arthus/fisiopatología , Linfocitos B/inmunología , Inmunoglobulina G/administración & dosificación , Proteína Estafilocócica A/administración & dosificación , Superantígenos/administración & dosificación , Animales , Reacción de Arthus/etiología , Linfocitos B/metabolismo , Femenino , Humanos , Inmunoglobulina G/inmunología , Inmunoglobulina G/metabolismo , Cadenas Pesadas de Inmunoglobulina/administración & dosificación , Cadenas Pesadas de Inmunoglobulina/inmunología , Región Variable de Inmunoglobulina/administración & dosificación , Región Variable de Inmunoglobulina/inmunología , Ratones , Ratones Endogámicos BALB C , Infiltración Neutrófila , Neutrófilos/inmunología , Cavidad Peritoneal/fisiopatología , Proteína Estafilocócica A/inmunología , Proteína Estafilocócica A/metabolismo , Superantígenos/inmunología , Superantígenos/metabolismo
15.
Infect Immun ; 73(9): 5540-6, 2005 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16113270

RESUMEN

We investigated the biological properties of a novel staphylococcal enterotoxin (SE)-like toxin type P (SElP). SElP induced a substantial proliferative response and the production of cytokines interleukin-2, gamma interferon, tumor necrosis factor alpha, and interleukin-4 from human T cells when administered at a concentration of 0.4 pM (0.01 ng/ml) or more. The expression of major histocompatibility complex class II molecules on accessory cells was required for T-cell stimulation by SElP. SElP selectively stimulated a vast number of human T cells bearing receptors Vbeta 5.1, 6, 8, 16, 18, and 21.3. These results indicated that SElP acts as a superantigen. SElP proved to be emetic in the house musk shrew emetic assay, although at a relatively high dose (50 to 150 mug/animal). A quantitative assay of SElP production with 30 Staphylococcus aureus strains harboring selp showed that 60% of these strains produced significant amounts of SElP in vitro. All 10 strains carrying seb and selp produced SEB but not SElP, suggesting the inactivation of the selp locus in S. aureus strains with a particular se gene constitution.


Asunto(s)
Enterotoxinas/química , Staphylococcus aureus/patogenicidad , Superantígenos/genética , Animales , Enterotoxinas/administración & dosificación , Enterotoxinas/genética , Enterotoxinas/inmunología , Humanos , Linfocitos/inmunología , Staphylococcus aureus/genética , Staphylococcus aureus/inmunología , Staphylococcus aureus/aislamiento & purificación , Superantígenos/administración & dosificación , Superantígenos/química , Superantígenos/inmunología
16.
J Immunol ; 173(10): 5944-51, 2004 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-15528328

RESUMEN

Mice deficient in OX40 or 4-1BB costimulatory pathways show defects in T cell recall responses, with predominant effects on CD4 vs CD8 T cells, respectively. However, OX40L can also stimulate CD8 T cells and 4-1BBL can influence CD4 T cells, raising the possibility of redundancy between the two TNFR family costimulators. To test this possibility, we generated mice deficient in both 4-1BBL and OX40L. In an adoptive transfer model, CD4 T cells expressed 4-1BB and OX40 sequentially in response to immunization, with little or no overlap in the timing of their expression. Under the same conditions, CD8 T cells expressed 4-1BB, but no detectable OX40. Thus, in vivo expression of 4-1BB and OX40 can be temporally and spatially segregated. In the absence of OX40L, there were decreased CD4 T cells late in the primary response and no detectable secondary expansion of adoptively transferred CD4 T cells under conditions in which primary expansion was unaffected. The 4-1BBL had a minor effect on the primary response of CD4 T cells in this model, but showed larger effects on the secondary response, although 4-1BBL(-/-) mice show less impairment in CD4 secondary responses than OX40L(-/-) mice. The 4-1BBL(-/-) and double knockout mice were similarly impaired in the CD8 T cell response, whereas OX40L(-/-) and double knockout mice were similarly impaired in the CD4 T cell response to both protein Ag and influenza virus. Thus, 4-1BB and OX40 act independently and nonredundantly to facilitate robust CD4 and CD8 recall responses.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Memoria Inmunológica , Receptores de Factor de Crecimiento Nervioso/fisiología , Receptores del Factor de Necrosis Tumoral/fisiología , Ligando 4-1BB , Traslado Adoptivo/métodos , Animales , Antígenos CD , Linfocitos T CD4-Positivos/microbiología , Linfocitos T CD4-Positivos/trasplante , Linfocitos T CD4-Positivos/virología , Linfocitos T CD8-positivos/microbiología , Linfocitos T CD8-positivos/trasplante , Linfocitos T CD8-positivos/virología , Proliferación Celular , Enterotoxinas/administración & dosificación , Enterotoxinas/inmunología , Epítopos de Linfocito T/inmunología , Inmunización Secundaria/métodos , Memoria Inmunológica/genética , Virus de la Influenza A/inmunología , Ligandos , Activación de Linfocitos/genética , Glicoproteínas de Membrana/deficiencia , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Glicoproteínas de Membrana/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Ligando OX40 , Receptores de Factor de Crecimiento Nervioso/biosíntesis , Receptores OX40 , Receptores del Factor de Necrosis Tumoral/biosíntesis , Superantígenos/administración & dosificación , Superantígenos/inmunología , Miembro 9 de la Superfamilia de Receptores de Factores de Necrosis Tumoral , Factor de Necrosis Tumoral alfa/deficiencia , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/metabolismo , Factor de Necrosis Tumoral alfa/fisiología , Factores de Necrosis Tumoral
17.
J Clin Oncol ; 22(4): 602-9, 2004 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-14966084

RESUMEN

PURPOSE: A patient-specific dose-escalation scheme using a Bayesian model of Escalation with Overdose Control (EWOC) was conducted to establish the maximum tolerated dose (MTD) of PNU-214936 in advanced non-small-cell lung cancer (NSCLC). PNU-214936 is a murine Fab fragment of the monoclonal antibody 5T4 fused to a mutated superantigen staphylococcal enterotoxin A (SEA). PATIENTS AND METHODS: Seventy-eight patients with NSCLC were treated with an individualized dose of PNU-214936 calculated using EWOC, based on their anti-SEA antibody level, and given as a 3-hour infusion on 4 consecutive days. RESULTS: Fever (82%; grade 3 to 4, 2.6%) and hypotension (57%; grade 3 to 4, 9%) were the most common toxicities. Eight dose-limiting toxicities occurred, as defined as any grade 4 toxicity occurring within the first 5 days. The MTD was defined as a function of pretreatment anti-SEA antibody level. MTD ranged from 103 ng/kg for patients with anti-SEA concentrations < or = 10 pmol/mL, to 601 ng/kg for patients with anti-SEA concentrations of 91 to 150 pmol/mL. A minor tumor response was demonstrated in five of 66 assessable patients. CONCLUSION: EWOC determined phase I doses of PNU-214936 that were adjusted for patient anti-SEA antibody level, while safeguarding against overdose. Furthermore, the method permitted the construction of a dosing algorithm that would allow patients in subsequent clinical investigations to be treated with a dose of PNU-214936 that is tailored to their specific tolerance for the agent, as reflected by their pretreatment anti-SEA.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Enterotoxinas/administración & dosificación , Inductores de Interferón/administración & dosificación , Neoplasias Pulmonares/tratamiento farmacológico , Superantígenos/administración & dosificación , Adulto , Anciano , Teorema de Bayes , Carcinoma de Pulmón de Células no Pequeñas/inmunología , Relación Dosis-Respuesta a Droga , Enterotoxinas/inmunología , Femenino , Humanos , Infusiones Intravenosas , Inductores de Interferón/inmunología , Neoplasias Pulmonares/inmunología , Masculino , Dosis Máxima Tolerada , Persona de Mediana Edad , Modelos Estadísticos , Superantígenos/inmunología
18.
J Immunol ; 171(7): 3475-84, 2003 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-14500643

RESUMEN

The repeated injection of low doses of bacterial superantigens (SAg) is known to induce specific T cell unresponsiveness. We show in this study that the spleen of BALB/c mice receiving chronically, staphylococcal enterotoxin B (SEB) contains SEB-specific CD4(+) TCRBV8(+) T cells exerting an immune regulatory function on SEB-specific primary T cell responses. Suppression affects IL-2 and IFN-gamma secretion as well as proliferation of T cells. However, the suppressor cells differ from the natural CD4(+) T regulatory cells, described recently in human and mouse, because they do not express cell surface CD25. They are CD152 (CTLA-4)-negative and their regulatory activity is not associated with expression of the NF Foxp3. By contrast, after repeated SEB injection, CD4(+)CD25(+) splenocytes were heterogenous and contained both effector as well as regulatory cells. In vivo, CD4(+)CD25(-) T regulatory cells prevented SEB-induced death independently of CD4(+)CD25(+) T cells. Nevertheless, SEB-induced tolerance could not be achieved in thymectomized CD25(+) cell-depleted mice because repeated injection of SEB did not avert lethal toxic shock in these animals. Collectively, these data demonstrate that, whereas CD4(+)CD25(+) T regulatory cells are required for the induction of SAg-induced tolerance, CD4(+)CD25(-) T cells exert their regulatory activity at the maintenance stage of SAg-specific unresponsiveness.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Anergia Clonal/inmunología , Enterotoxinas/administración & dosificación , Receptores de Interleucina-2/biosíntesis , Superantígenos/administración & dosificación , Subgrupos de Linfocitos T/inmunología , Linfocitos T Reguladores/inmunología , Animales , Antígenos CD , Antígenos de Diferenciación/biosíntesis , Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD4-Positivos/microbiología , Antígeno CTLA-4 , Movimiento Celular/inmunología , Separación Celular , Relación Dosis-Respuesta Inmunológica , Regulación hacia Abajo/inmunología , Esquema de Medicación , Enterotoxinas/inmunología , Epítopos de Linfocito T/administración & dosificación , Epítopos de Linfocito T/inmunología , Femenino , Inyecciones Intraperitoneales , Depleción Linfocítica , Ratones , Ratones Endogámicos BALB C , Bazo/citología , Bazo/inmunología , Staphylococcus aureus/inmunología , Superantígenos/inmunología , Subgrupos de Linfocitos T/metabolismo , Subgrupos de Linfocitos T/microbiología , Linfocitos T Reguladores/metabolismo , Linfocitos T Reguladores/microbiología , Timo/citología , Timo/inmunología
19.
J Immunol ; 170(10): 5008-17, 2003 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-12734345

RESUMEN

Repeated exposures to both microbial and innocuous Ags in vivo have been reported to both eliminate and tolerize T cells after their initial activation and expansion. The remaining tolerant T cells have been shown to suppress the response of naive T cells in vitro. This feature is reminiscent of natural CD4(+)CD25(+) regulatory T cells. However, it is not known whether the regulatory function of in vivo-tolerized T cells is similar to the function of natural CD4(+)CD25(+) regulatory T cells. In this study, we demonstrate that CD4(+)CD25(+) as well as CD4(+)CD25(-) T cells isolated from mice treated with superantigen three consecutive times to induce tolerance were functionally comparable to natural CD4(+)CD25(+) regulatory T cells, albeit more potent. The different subpopulations of in vivo-tolerized CD4(+) T cells efficiently down-modulated costimulatory molecules on dendritic cells, and their suppressive functions were strictly cell contact dependent. Importantly, we demonstrate that conventional CD4(+)CD25(-) T cells could also be induced to acquire regulatory functions by the same regimen in the absence of natural regulatory T cells in vivo, but that such regulatory cells were functionally different.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Regulación hacia Abajo/inmunología , Enterotoxinas/administración & dosificación , Inmunoconjugados , Activación de Linfocitos/inmunología , Receptores de Interleucina-2/biosíntesis , Superantígenos/administración & dosificación , Subgrupos de Linfocitos T/inmunología , Abatacept , Animales , Células Presentadoras de Antígenos/inmunología , Células Presentadoras de Antígenos/metabolismo , Antígenos CD , Antígenos de Diferenciación/biosíntesis , Linfocitos T CD4-Positivos/metabolismo , Antígeno CTLA-4 , Comunicación Celular/genética , Comunicación Celular/inmunología , Células Cultivadas , Anergia Clonal/genética , Anergia Clonal/inmunología , Supresión Clonal/genética , Supresión Clonal/inmunología , Citocinas/fisiología , Proteínas de Unión al ADN/biosíntesis , Proteínas de Unión al ADN/deficiencia , Proteínas de Unión al ADN/genética , Relación Dosis-Respuesta Inmunológica , Regulación hacia Abajo/genética , Esquema de Medicación , Enterotoxinas/farmacología , Femenino , Genes Codificadores de la Cadena beta de los Receptores de Linfocito T/inmunología , Inmunidad Innata/genética , Inyecciones Intravenosas , Interleucina-10/antagonistas & inhibidores , Interleucina-10/metabolismo , Antígenos Comunes de Leucocito/biosíntesis , Activación de Linfocitos/genética , Ratones , Ratones Endogámicos C57BL , Ratones SCID , Ratones Transgénicos , Staphylococcus aureus/inmunología , Superantígenos/farmacología , Subgrupos de Linfocitos T/metabolismo , Factor de Crecimiento Transformador beta/antagonistas & inhibidores , Factor de Crecimiento Transformador beta/metabolismo , Factor de Crecimiento Transformador beta/fisiología
20.
Crit Care Med ; 31(4): 1154-9, 2003 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-12682487

RESUMEN

OBJECTIVE: To determine the effect of chronic exposure to endotoxin (lipopolysaccharide) and Staphylococcal enterotoxin B on hepatic injury and function. DESIGN: Prospective, controlled trial. SETTING: Research laboratory in a university hospital. SUBJECTS: Male Sprague-Dawley rats weighing 325-350 g with chronic vascular and bile catheters. INTERVENTIONS: Chronically catheterized rats were treated daily with saline, 50 microg/kg Staphylococcal enterotoxin B alone, 1000 microg/kg lipopolysaccharide alone, 1000 microg/kg lipopolysaccharide with 50 microg/kg Staphylococcal enterotoxin B, or 100 microg/kg lipopolysaccharide with 50 microg/kg Staphylococcal enterotoxin B for 10 days. Serum and biliary measures of hepatic injury and dysfunction were measured before and then 6 hrs and 1, 2, 3, 7, and 10 days after the start of treatment. The animals were killed at 10 days and the livers examined histologically. MEASUREMENTS AND MAIN RESULTS: Mean rates of bile flow, biliary indocyanine green excretion, and bile acid flux were significantly decreased immediately after treatment (6 hr, 1 and 2 days) and then at 10 days. Increases in biliary and serum gamma-glutamyltransferase and serum bile acids also occurred in a similar bimodal pattern. Animals treated with lipopolysaccharide or Staphylococcal enterotoxin B alone became tolerant and did not develop the bimodal pattern of hepatic dysfunction. Histologic examination of the liver at 10 days revealed periportal inflammation and fibrosis. CONCLUSIONS: The combination of lipopolysaccharide and Staphylococcal enterotoxin B leads to late liver injury, whereas either toxin alone does not. These data may explain the frequent development of liver dysfunction in patients exposed to multiple bacterial toxins such as in sepsis, multiple-system organ failure, and other diseases with altered intestinal permeability.


Asunto(s)
Enterotoxinas/administración & dosificación , Lipopolisacáridos/administración & dosificación , Hepatopatías/fisiopatología , Hígado/fisiopatología , Sepsis/fisiopatología , Staphylococcus aureus , Superantígenos/administración & dosificación , Alanina Transaminasa/sangre , Animales , Bilis/fisiología , Ácidos y Sales Biliares/sangre , Escherichia coli , Verde de Indocianina , Infusiones Intravenosas , Interferón gamma/sangre , Hepatopatías/etiología , Pruebas de Función Hepática , Masculino , Insuficiencia Multiorgánica/fisiopatología , Ratas , Ratas Sprague-Dawley , Sepsis/complicaciones , Factor de Necrosis Tumoral alfa/análisis , gamma-Glutamiltransferasa/análisis
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA