Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
FASEB J ; 38(7): e23569, 2024 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-38551610

RESUMEN

Early in sepsis, a hyperinflammatory response is dominant, but later, an immunosuppressive phase dominates, and the host is susceptible to opportunistic infections. Anti-inflammatory agents may accelerate the host into immunosuppression, and few agents can reverse immunosuppression without causing inflammation. Specialized pro-resolving mediators (SPMs) such as resolvin D2 (RvD2) have been reported to resolve inflammation without being immunosuppressive, but little work has been conducted to examine their effects on immunosuppression. To assess the effects of RvD2 on immunosuppression, we established a model of macrophage exhaustion using two lipopolysaccharide (LPS) treatments or hits. THP-1 monocyte-derived macrophages were first treated with RvD2 or vehicle for 1 h. One LPS hit increased NF-κB activity 11-fold and TNF-α release 60-fold compared to unstimulated macrophages. RvD2 decreased LPS-induced NF-κB activity and TNF-α production but increased bacterial clearance. Two LPS hits reduced macrophage bacterial clearance and decreased macrophage NF-κB activity (45%) and TNF-α release (75%) compared to one LPS hit, demonstrating exhaustion. RvD2 increased NF-κB activity, TNF-α release, and bacterial clearance following two LPS hits compared to controls. TLR2 inhibition abolished RvD2-mediated changes. In a mouse sepsis model, splenic macrophage response to exogenous LPS was reduced compared to controls and was restored by in vivo administration of RvD2, supporting the in vitro results. If RvD2 was added to monocytes before differentiation into macrophages, however, RvD2 reduced LPS responses and increased bacterial clearance following both one and two LPS hits. The results show that RvD2 attenuated macrophage suppression in vitro and in vivo and that this effect was macrophage-specific.


Asunto(s)
Ácidos Docosahexaenoicos , Lipopolisacáridos , Sepsis , Ratones , Animales , Lipopolisacáridos/toxicidad , FN-kappa B/farmacología , Factor de Necrosis Tumoral alfa/farmacología , Macrófagos , Inflamación/inducido químicamente , Inflamación/tratamiento farmacológico , Sepsis/inducido químicamente , Sepsis/tratamiento farmacológico
2.
Biomedicines ; 12(1)2024 Jan 13.
Artículo en Inglés | MEDLINE | ID: mdl-38255280

RESUMEN

Severe infection can lead to sepsis. In sepsis, the host mounts an inappropriately large inflammatory response in an attempt to clear the invading pathogen. This sustained high level of inflammation may cause tissue injury and organ failure. Later in sepsis, a paradoxical immunosuppression occurs, where the host is unable to clear the preexisting infection and is susceptible to secondary infections. A major issue with sepsis treatment is that it is difficult for physicians to ascertain which stage of sepsis the patient is in. Sepsis treatment will depend on the patient's immune status across the spectrum of the disease, and these immune statuses are nearly polar opposites in the early and late stages of sepsis. Furthermore, there is no approved treatment that can resolve inflammation without contributing to immunosuppression within the host. Here, we review the major mechanisms of sepsis-induced immunosuppression and the biomarkers of the immunosuppressive phase of sepsis. We focused on reviewing three main mechanisms of immunosuppression in sepsis. These are lymphocyte apoptosis, monocyte/macrophage exhaustion, and increased migration of myeloid-derived suppressor cells (MDSCs). The biomarkers of septic immunosuppression that we discuss include increased MDSC production/migration and IL-10 levels, decreased lymphocyte counts and HLA-DR expression, and increased GPR18 expression. We also review the literature on the use of specialized pro-resolving mediators (SPMs) in different models of infection and/or sepsis, as these compounds have been reported to resolve inflammation without being immunosuppressive. To obtain the necessary information, we searched the PubMed database using the keywords sepsis, lymphocyte apoptosis, macrophage exhaustion, MDSCs, biomarkers, and SPMs.

3.
FASEB J ; 37(8): e23098, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37462621

RESUMEN

Pseudomonas aeruginosa is a gram-negative, opportunistic bacteria commonly found in wounds and in lungs of immunocompromised patients. These bacteria commonly form biofilms which encapsulate the bacteria, making it difficult for antibiotics or immune cells to reach the bacterial cells. We previously reported that Lipoxin A4 (LxA4 ), a Specialized Pro-resolving Mediator, has direct effects on P. aeruginosa where it reduced biofilm formation and promoted ciprofloxacin antibiotic efficacy in a static biofilm-forming system. In the current studies, we examined the actions of LxA4 on established biofilms formed in a biofilm reactor under dynamic conditions with constant flow and shear stress. These conditions allow for biofilm growth with nutrient replenishment and for examination of bacteria within the biofilm structure. We show that LxA4 helped ciprofloxacin reduction of live/dead ratio of bacteria within the biofilm. THP-1 monocytes interacted with the biofilm to increase the number of viable bacteria within the biofilm as well as TNF-α production in the biofilm milieu, suggesting that monocyte interaction with bacterial biofilm exacerbates the inflammatory state. Pre-treatment of the THP-1 monocytes with LxA4 abolished the increase in biofilm bacteria and reduced TNF-α production. The effect of decreased biofilm bacteria was associated with increased LxA4 -induced monocyte adherence to biofilm but not increased bacteria killing suggesting that the mechanism for the reduced biofilm bacteria was due to LxA4 -mediated increase in adherence to biofilm. These results suggest that LxA4 can help antibiotic efficacy and promote monocyte activity against established P. aeruginosa biofilm formed under hydrodynamic conditions.


Asunto(s)
Lipoxinas , Monocitos , Humanos , Antibacterianos/farmacología , Pseudomonas aeruginosa , Lipoxinas/farmacología , Hidrodinámica , Factor de Necrosis Tumoral alfa/farmacología , Biopelículas , Ciprofloxacina/farmacología
4.
Front Immunol ; 13: 1011944, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36532055

RESUMEN

In severe bacterial infections, there is a pro-inflammatory response to promote bacterial clearance but this response can cause tissue injury. Later, the immune system becomes dysregulated and the host is unable to clear a secondary or a pre-existing infection. Specialized Pro-resolving Mediators (SPMs) such as resolvin D2 (RvD2) have been shown to be beneficial for inflammation/infection resolution in animal models of sepsis but in vivo mechanisms by which RvD2 may promote bacterial clearance and/or attenuate deleterious effects of a secondary infection have not been fully established. In this study, we used the 2-hit model of cecal ligation and puncture (CLP) induced infectious peritonitis and secondary lung infection with Pseudomonas aeruginosa to find possible antimicrobial and immunomodulatory mechanisms of RvD2. We show that RvD2 given as late as 48h after CLP surgery reduced blood bacterial load without altering plasma cytokines compared to mice given saline vehicle. RvD2 increased splenic neutrophil accumulation as well as average reactive oxygen species (ROS) production. There was also an increase in an immature leukocyte population the myeloid derived suppressor cells (MDSCs) in the spleen of RvD2 treated mice. RvD2 reduced lung lavage bacterial load 24h after P. aeruginosa administration and significantly decreased lung lavage levels of IL-23, a cytokine essential in the Th-17 inflammatory response. In addition, we show that RvD2 increased the number of non-inflammatory alveolar macrophages after P. aeruginosa administration compared to saline treated mice. The study uncovered an antimicrobial mechanism of RvD2 where RvD2 increases mature neutrophil and MDSC accumulation into the spleen to promote blood bacterial clearance. The study showed that in this 2-hit model, RvD2 promotes lung bacterial clearance, increased non-inflammatory alveolar macrophage number and inhibits an adaptive immune pathway providing evidence of its resolution mechanism in secondary pulmonary infection.


Asunto(s)
Coinfección , Peritonitis , Neumonía , Ratones , Animales , Coinfección/metabolismo , Modelos Animales de Enfermedad , Pulmón , Peritonitis/tratamiento farmacológico , Peritonitis/metabolismo , Citocinas/metabolismo , Pseudomonas aeruginosa , Neumonía/tratamiento farmacológico , Neumonía/etiología , Neumonía/metabolismo
5.
Molecules ; 26(22)2021 Nov 18.
Artículo en Inglés | MEDLINE | ID: mdl-34834062

RESUMEN

Bacterial infection activates the innate immune system as part of the host's defense against invading pathogens. Host response to bacterial pathogens includes leukocyte activation, inflammatory mediator release, phagocytosis, and killing of bacteria. An appropriate host response requires resolution. The resolution phase involves attenuation of neutrophil migration, neutrophil apoptosis, macrophage recruitment, increased phagocytosis, efferocytosis of apoptotic neutrophils, and tissue repair. Specialized Pro-resolving Mediators (SPMs) are bioactive fatty acids that were shown to be highly effective in promoting resolution of infectious inflammation and survival in several models of infection. In this review, we provide insight into the role of SPMs in active host defense mechanisms for bacterial clearance including a new mechanism of action in which an SPM acts directly to reduce bacterial virulence.


Asunto(s)
Bacterias/inmunología , Infecciones Bacterianas/inmunología , Mediadores de Inflamación/inmunología , Inflamación/inmunología , Fagocitosis , Animales , Bacterias/patogenicidad , Infecciones Bacterianas/complicaciones , Ácidos Docosahexaenoicos/inmunología , Humanos , Inmunidad Innata , Inflamación/complicaciones , Lipoxinas/inmunología , Macrófagos/inmunología , Neutrófilos/inmunología , Virulencia
6.
Front Cell Infect Microbiol ; 11: 737392, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34589444

RESUMEN

Probiotics such as various strains of Lactobacillaceae have been shown to have antimicrobial and immunomodulatory activity. In vitro studies have shown that Lactobacilli can decrease bacterial biofilm formation. Effects on immune cells have been unclear with most studies showing anti-inflammatory activity. The mechanism of effects has not been clearly elucidated. In these studies, we used different concentrations of live Lactobacillus acidophilus as well as cell free filtrate (CFF) derived from different concentrations of bacteria. Use of CFF is advantageous as a therapeutic because in vivo it can directly contact immune cells and its concentration is fixed. Both live cells and CFF inhibited Pseudomonas aeruginosa biofilm formation. Importantly, we show that high concentration CFF destroyed mature biofilm. This activity was not due to a lowered pH per se, as pH matched HCl did not remove mature biofilm. High concentration CFF totally inhibited P. aeruginosa growth and was bactericidal (>99.99%), but low concentration CFF was not bactericidal. To examine the immunomodulatory effects of L. acidophilus, we incubated THP-1 monocytes and derived macrophages with CFF and measured TNFα production. CFF did not significantly increase TNFα production in THP-1 monocytes. When cells were prestimulated with LPS, high concentration CFF increased TNFα production even further. In macrophages, high concentration CFF alone increased TNFα production but did not affect LPS prestimulated cells. In contrast, low concentration CFF decreased TNFα production in LPS prestimulated cells. To elucidate the possible mechanisms for these effects, we repeated the experiments using a NF-κB reporter THP-1 cell line. High concentration CFF increased NF-κB activity in monocytes and macrophages. In LPS prestimulated macrophages, only low concentration CFF reduced NF-κB activity. These results suggest that high concentration CFF alone induced NF-κB expression which could account partially for an increase in TNFα production. On the other hand, in macrophages, the lower non-bactericidal concentration of CFF reduced NF-κB expression and decreased TNFα production after LPS prestimulation. Taken together, the results provide evidence that different concentrations of L. acidophilus CFF possess varying bactericidal, anti-biofilm and immunomodulatory effects. This is important in vivo to evaluate the possible use of L. acidophilus CFF in different conditions.


Asunto(s)
Lactobacillus acidophilus , Probióticos , Biopelículas , Lipopolisacáridos , Monocitos , FN-kappa B , Factor de Necrosis Tumoral alfa
7.
FASEB J ; 30(6): 2400-10, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-26965685

RESUMEN

Bacterial infections can quickly turn into sepsis, with its attendant clinical sequelae of inflammation, tissue injury, and organ failure. Paradoxically, sustained inflammation in sepsis may lead to immune suppression, because of which the host is unable to clear the existing infection. Use of agents that suppress the inflammatory response may accelerate host immune suppression, whereas use of traditional antibiotics does not significantly affect inflammation. In this study, we investigated whether lipoxin A4 (LXA4), a specialized, proresolution lipid mediator, could increase neutrophil phagocytic activity as well as reduce bacterial virulence. Using the mouse cecal ligation and puncture (CLP) model of sepsis, the administration of LXA4 (7 µg/kg i.v.) 1 h after surgery increased neutrophil phagocytic ability and Fcγ receptor I (CD64) expression. Ex vivo studies have confirmed that the direct addition of LXA4 to CLP neutrophils increased phagocytic ability but not CD64 expression. LXA4 did not affect neutrophils taken from control mice in which CD64 expression was minimal. Taken together with in vivo data, these results suggest that LXA4 directly augments CD64-mediated neutrophil phagocytic ability but does not directly increase neutrophil CD64 expression. Bacterial communication and virulence is regulated by quorum sensing inducers. In Pseudomonas aeruginosa, virulence is induced with release of various virulence factors, by N-3-oxododecanolyl homoserine lactone binding to the quorum sensing receptor, LasR. We show that LXA4 is an inhibitor of LasR in P. aeruginosa and that it decreases the release of pyocyanin exotoxin. These results suggest that LXA4 has the novel dual properties of increasing host defense and decreasing pathogen virulence by inhibiting quorum sensing.-Wu, B., Capilato, J., Pham, M. P., Walker, J., Spur, B., Rodriguez, A., Perez, L. J., Yin, K. Lipoxin A4 augments host defense in sepsis and reduces Pseudomonas aeruginosa virulence through quorum sensing inhibition.


Asunto(s)
Lipoxinas/metabolismo , Infecciones por Pseudomonas/inmunología , Pseudomonas aeruginosa/patogenicidad , Percepción de Quorum/efectos de los fármacos , Sepsis/inmunología , Animales , Anticuerpos/fisiología , Bacteriemia , Proteínas Bacterianas , Leucocitos/efectos de los fármacos , Lipoxinas/genética , Masculino , Ratones , Neutrófilos , Fagocitos , Infecciones por Pseudomonas/microbiología , Receptores de IgG/genética , Receptores de IgG/metabolismo , Sepsis/metabolismo , Transactivadores , Virulencia
8.
Oncotarget ; 6(11): 9045-60, 2015 Apr 20.
Artículo en Inglés | MEDLINE | ID: mdl-25826087

RESUMEN

Podoplanin (PDPN) is a unique transmembrane receptor that promotes tumor cell motility. Indeed, PDPN may serve as a chemotherapeutic target for primary and metastatic cancer cells, particularly oral squamous cell carcinoma (OSCC) cells that cause most oral cancers. Here, we studied how a monoclonal antibody (NZ-1) and lectin (MASL) that target PDPN affect human OSCC cell motility and viability. Both reagents inhibited the migration of PDPN expressing OSCC cells at nanomolar concentrations before inhibiting cell viability at micromolar concentrations. In addition, both reagents induced mitochondrial membrane permeability transition to kill OSCC cells that express PDPN by caspase independent nonapoptotic necrosis. Furthermore, MASL displayed a surprisingly robust ability to target PDPN on OSCC cells within minutes of exposure, and significantly inhibited human OSCC dissemination in zebrafish embryos. Moreover, we report that human OSCC cells formed tumors that expressed PDPN in mice, and induced PDPN expression in infiltrating host murine cancer associated fibroblasts. Taken together, these data suggest that antibodies and lectins may be utilized to combat OSCC and other cancers that express PDPN.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Antineoplásicos Fitogénicos/farmacología , Carcinoma de Células Escamosas/patología , Glicoproteínas de Membrana/antagonistas & inhibidores , Terapia Molecular Dirigida , Neoplasias de la Boca/patología , Proteínas de Neoplasias/antagonistas & inhibidores , Fitohemaglutininas/farmacología , Administración Oral , Animales , Animales Modificados Genéticamente , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales/uso terapéutico , Antineoplásicos Fitogénicos/administración & dosificación , Antineoplásicos Fitogénicos/uso terapéutico , Carcinoma de Células Escamosas/virología , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Fibroblastos/patología , Humanos , Glicoproteínas de Membrana/inmunología , Glicoproteínas de Membrana/fisiología , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Ratones , Neoplasias de la Boca/virología , Proteínas de Neoplasias/inmunología , Proteínas de Neoplasias/fisiología , Papillomaviridae/aislamiento & purificación , Infecciones por Papillomavirus/patología , Infecciones por Papillomavirus/virología , Fitohemaglutininas/administración & dosificación , Fitohemaglutininas/uso terapéutico , Ensayos Antitumor por Modelo de Xenoinjerto , Pez Cebra/embriología
9.
Shock ; 36(4): 410-6, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21701419

RESUMEN

Sepsis is characterized by systemic inflammation with release of a large amount of inflammatory mediators. If sustained, this inflammatory response can lead to multiple organ failure and/or immunoparalysis. In the latter condition, the host may be susceptible to opportunistic infections or be unable to clear existing infections. Therefore, it is potentially beneficial to resolve inflammation by reducing inflammation without compromising host defense. We examined the effect of lipoxin A4 (LXA4), a compound with inflammatory resolution properties, in the cecal ligation and puncture (CLP) model of sepsis. Cecal ligation and puncture rats were given either saline or LXA4 (40 µg/kg, i.p.) 5 h after surgery. Lipoxin A4 administration increased 8-day survival of CLP rats, which lived longer than 48 h, and attenuated tissue injury after 8 days. Therefore, we investigated the effects of LXA4 on systemic inflammation and bacterial load 48 h after CLP sepsis. Plasma IL-6, monocyte chemotactic protein 1, and IL-10 levels were reduced in LXA4-treated rats compared with CLP rats given saline vehicle. Lipoxin A4 reduced phosphorylation of the p65 subunit of nuclear factor κB (NF-κB) at serines 536 and 468 in peritoneal macrophages, suggesting that LXA4 reduced production of proinflammatory mediators through an NF-κB-mediated mechanism. Lipoxin A4 reduced blood bacterial load and increased peritoneal macrophage number without affecting phagocytic ability, suggesting that LXA4 reduced blood bacterial load by enhancing macrophage recruitment. It also suggests that LXA4 reduced systemic inflammation and NF-κB activation without compromising host defense. Increased macrophage recruitment was in part due to a direct effect of LXA4 as LXA4 increased peritoneal macrophage recruitment in sham controls and partly due to reduced production of IL-10 as LXA4 decreased macrophage IL-10 release (a known inhibitor of macrophage migration) after CLP. The results suggest that LXA4 increased survival in sepsis by simultaneously reducing systemic inflammation as well as bacterial spread.


Asunto(s)
Lipoxinas/uso terapéutico , Sepsis/tratamiento farmacológico , Sepsis/inmunología , Animales , Carga Bacteriana , Quimiocina CCL2/metabolismo , Inflamación/tratamiento farmacológico , Inflamación/microbiología , Interleucina-10/metabolismo , Interleucina-6/metabolismo , Estimación de Kaplan-Meier , Masculino , FN-kappa B/metabolismo , Fagocitosis/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Sepsis/microbiología
10.
BMC Genomics ; 9: 608, 2008 Dec 16.
Artículo en Inglés | MEDLINE | ID: mdl-19087328

RESUMEN

BACKGROUND: Lipopolysaccharide (LPS) is a cell wall component of Gram-negative bacteria with proved role in pathogenesis of sepsis. Brain injury was observed with both patients dead from sepsis and animal septic models. However, in vitro administration of LPS has not shown obvious cell damage to astrocytes and other relative cell lines while it does cause endothelial cell death in vitro. These observations make it difficult to understand the role of LPS in brain parenchymal injury. RESULTS: To test the hypothesis that LPS may cause biological changes in astrocytes and make the cells to become vulnerable to reactive oxygen species, a recently developed highly sensitive and highly specific system for large-scale gene expression profiling was used to examine the gene expression profile of a group of 1,135 selected genes in a cell line, T98G, a derivative of human glioblastoma of astrocytic origin. By pre-treating T98G cells with different dose of LPS, it was found that LPS treatment caused a broad alteration in gene expression profile, but did not cause obvious cell death. However, after short exposure to H2O2, cell death was dramatically increased in the LPS pretreated samples. Interestingly, cell death was highly correlated with down-regulated expression of antioxidant genes such as cytochrome b561, glutathione s-transferase a4 and protein kinase C-epsilon. On the other hand, expression of genes encoding growth factors was significantly suppressed. These changes indicate that LPS treatment may suppress the anti-oxidative machinery, decrease the viability of the T98G cells and make the cells more sensitive to H2O2 stress. CONCLUSION: These results provide very meaningful clue for further exploring and understanding the mechanism underlying astrocyte injury in sepsis in vivo, and insight for why LPS could cause astrocyte injury in vivo, but not in vitro. It will also shed light on the therapeutic strategy of sepsis.


Asunto(s)
Antioxidantes/metabolismo , Astrocitos/metabolismo , Peróxido de Hidrógeno/toxicidad , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Lipopolisacáridos/farmacología , Astrocitos/efectos de los fármacos , Astrocitoma , Muerte Celular , Línea Celular Tumoral , Expresión Génica , Perfilación de la Expresión Génica , Humanos , Péptidos y Proteínas de Señalización Intercelular/genética , Oxidación-Reducción , Estrés Oxidativo , Sepsis/metabolismo
11.
Shock ; 29(6): 754-60, 2008 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-18004227

RESUMEN

Macrophage dysfunction occurs late in sepsis and is implicated in increased mortality. Interferon gamma (IFN-gamma) stimulates transmigration of high mobility group box 1 (HMGB-1) from the nucleus into cytoplasm of macrophages and subsequent release. Because HMGB-1 release also occurs late, and because one of the actions of HMGB-1 in the nucleus is to enhance transcription factors, we investigated if HMGB-1 transmigration is involved in macrophage suppression in sepsis. Alveolar macrophages were isolated 12 and 24 h from sham controls, cecal ligation and puncture (CLP), and CLP rats given IFN-gamma antibody (1.2 mg/kg, i.v.). All injections were given immediately after surgery. At 12 h, 60% of cells from sham controls had HMGB-1 located primarily in the nucleus, whereas 35% of cells had diffuse staining in both cytoplasm and nucleus. In CLP rats, HMGB-1 was located predominantly in the cytoplasm of 37% of cells, and 48% had diffuse staining, whereas in IFN-gamma antibody (Ab)-treated rats, HMGB-1 was located predominantly in the nucleus of 56% of cells, whereas 32% had diffuse staining. At 24 h, most cells from CLP rats (82%) had HMGB-1 located in the cytoplasm, whereas in contrast, HMGB-1 was located in the nucleus of 80% and 82% of cells from sham control and IFN-gamma Ab-treated rats, respectively. Gene expression of TNF-alpha was not significantly changed 12 h after surgery, but at 24 h, alveolar macrophages from CLP rats had reduced gene expression of TNF-alpha. Interferon gamma Ab treatment prevented the reduction in TNF-alpha gene expression. TNF-alpha release was not altered at 12 h. At 24 h, LPS-stimulated release of TNF-alpha was decreased in macrophages from CLP rats compared with sham controls. Interferon gamma Ab treatment prevented the decrease in LPS-stimulated TNF-alpha release. The results suggest that alveolar macrophage suppression after CLP is associated with HMGB-1 transmigration out of the cell nucleus and provides evidence that intranuclear HMGB-1 may play an integral role in macrophage activation in sepsis.


Asunto(s)
Núcleo Celular/metabolismo , Citoplasma/metabolismo , Proteínas del Grupo de Alta Movilidad/metabolismo , Activación de Macrófagos , Macrófagos Alveolares/metabolismo , Proteínas Represoras/metabolismo , Sepsis/metabolismo , Transporte Activo de Núcleo Celular/efectos de los fármacos , Animales , Anticuerpos/farmacología , Núcleo Celular/patología , Citoplasma/patología , Regulación de la Expresión Génica/efectos de los fármacos , Proteína HMGB1 , Interferón gamma/antagonistas & inhibidores , Interferón gamma/metabolismo , Lipopolisacáridos/farmacología , Activación de Macrófagos/efectos de los fármacos , Macrófagos Alveolares/patología , Masculino , Ratas , Ratas Sprague-Dawley , Sepsis/patología , Factores de Tiempo , Factor de Necrosis Tumoral alfa/biosíntesis
12.
J Surg Res ; 137(1): 16-20, 2007 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17112543

RESUMEN

BACKGROUND: One lung ventilation is a commonly performed surgical procedure. Although there have been several reports showing that one-lung ventilation can cause pathophysiological alterations such as pulmonary hypoxic vasoconstriction and intrapulmonary shunting, there have been virtually no reports on the effects of one-lung ventilation on lung histology. MATERIALS AND METHODS: Yorkshire pigs (11-17 kg) were anesthetized, a tracheotomy performed and a tracheal tube inserted. The chest was opened and one lung ventilation (OLV), was induced by clamping of the right main bronchus. OLV was continued for 60 min before the clamp was removed and two lung ventilation (TLV) started. TLV was continued for 30 to 60 min. Blood and lung biopsies were taken immediately before OLV, 30 min and 60 min of OLV and after restoration of TLV. RESULTS: Histological analyses revealed that the non-ventilated lung was totally collapsed during OLV. On reventilation, there was clear evidence of vascular congestion and alveolar wall thickening at 30 min after TLV. At 60 min of TLV, there was still vascular congestion. Serum nitrite levels (as an index of nitric oxide production) showed steady decline over the course of the experimental period, reaching a significantly low level on reventilation (compared with baseline levels before OLV). Lung MPO activity (marker of neutrophil sequestration) and serum TNFalpha levels were not raised during the entire experimental period. CONCLUSIONS: These results suggest that there was lung vascular injury after OLV, which was associated with reduced levels of nitric oxide production and not associated with an inflammatory response.


Asunto(s)
Pulmón/patología , Pulmón/cirugía , Complicaciones Posoperatorias/patología , Atelectasia Pulmonar/patología , Respiración Artificial/métodos , Animales , Pulmón/inmunología , Neutrófilos/enzimología , Nitritos/sangre , Oxígeno/sangre , Peroxidasa/metabolismo , Complicaciones Posoperatorias/inmunología , Complicaciones Posoperatorias/fisiopatología , Atelectasia Pulmonar/inmunología , Atelectasia Pulmonar/fisiopatología , Circulación Pulmonar , Sus scrofa , Factor de Necrosis Tumoral alfa/sangre , Vasoconstricción
13.
Shock ; 24(4): 396-401, 2005 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-16205327

RESUMEN

Interferon (IFN)-gamma is an important immunomodulatory agent that is stimulated during infection to aid in host defense. However, increased IFN-gamma levels have been implicated as a mediator in various models of tissue injury and endotoxemia. We have previously shown that inhibition of IFN-gamma decreased bacterial load by accelerating peritoneal fibrin deposition in the cecal ligation and puncture (CLP) model of peritonitis. In addition, circulating inflammatory mediators such as interleukin (IL)-6 were reduced by IFN-gamma inhibition. In the present study, we show that administration of IFN-gamma antibody (1.2 mg/kg, i.v.) attenuated mortality after CLP. Administration of this antibody was able to reduce mortality when given immediately after CLP or 24 h after CLP surgery. Mortality in sepsis has been closely associated with increased release of high mobility group box-1 (HMGB1). Furthermore, it has been reported that IFN-gamma stimulates the release of HMGB1 from macrophages. Our studies showed that inhibition of IFN-gamma activity in vivo reduced the levels of HMGB1 in peritoneal fluid and serum of CLP rats 24 h after surgery. In addition, the decrease in HMGB1 was associated with an increase in tissue repair as evidenced by histological analyses. These results suggest that the attenuation of mortality in IFN-gamma antibody-treated rats was associated with a decrease in HMGB1 release.


Asunto(s)
Proteína HMGB1/química , Interferón gamma/antagonistas & inhibidores , Sepsis/tratamiento farmacológico , Animales , Western Blotting , Ciego/patología , Inflamación , Interferón gamma/química , Interferón gamma/metabolismo , Ligadura , Masculino , Ratas , Ratas Sprague-Dawley , Sepsis/mortalidad , Factores de Tiempo , Cicatrización de Heridas
14.
Infect Immun ; 71(5): 2766-74, 2003 May.
Artículo en Inglés | MEDLINE | ID: mdl-12704151

RESUMEN

Bowel perforation can lead to significant bacterial spillage, which may then cause septic peritonitis, characterized by a systemic inflammatory response and organ dysfunction. There are several reports that have shown that the development of peritoneal adhesions is dependent on inflammatory cytokine levels and that these adhesions can reduce bacterial spread, possibly by sealing off the cecum in the cecal ligation and puncture (CLP) model of septic peritonitis. There have not, however, been any studies that have utilized a strategy to accelerate tissue repair in order to seal off the injured cecum and reduce bacterial spread as well as ameliorate systemic inflammation. In the present study, we demonstrate that the administration of anti-gamma interferon (IFN-gamma) antibody (1.2 mg/kg of body weight, intravenously) accelerated tissue repair via increased fibrin deposition 12 and 24 h after CLP in rats. This increase in fibrin deposition was associated with peritoneal adhesion 24 h after CLP and a reduction in bacterial load compared to the bacterial load of rats given irrelevant antibody. Plasma fibrin levels, however, were not altered after IFN-gamma antibody administration, suggesting that the inhibition of IFN-gamma activity specifically increased fibrin deposition to the site of injury. Furthermore, plasma interleukin-6, used as a marker of systemic inflammatory response, was reduced in CLP rats given IFN-gamma antibody compared to that found in those given irrelevant antibody. These results suggest that the early inhibition of IFN-gamma activity in the CLP model is beneficial by accelerating fibrin deposition in cecal tissue to prevent bacterial spread and reduce the systemic inflammatory response. Importantly, increased fibrin deposition in the ceca was not associated with increased plasma fibrin whereas the latter may have detrimental effects associated with coagulation disorders.


Asunto(s)
Bacterias/aislamiento & purificación , Fibrina/metabolismo , Interferón gamma/antagonistas & inhibidores , Peritonitis/microbiología , Animales , Western Blotting , Fibrinógeno/metabolismo , Interferón gamma/sangre , Interleucina-6/sangre , Masculino , Inhibidor 1 de Activador Plasminogénico/análisis , Ratas , Ratas Sprague-Dawley
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA