Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
1.
Front Immunol ; 15: 1374818, 2024.
Article in English | MEDLINE | ID: mdl-38827738

ABSTRACT

Activated lung ILC2s produce large quantities of IL-5 and IL-13 that contribute to eosinophilic inflammation and mucus production following respiratory syncytial virus infection (RSV). The current understanding of ILC2 activation during RSV infection, is that ILC2s are activated by alarmins, including IL-33, released from airway epithelial cells in response to viral-mediated damage. Thus, high levels of RSV neutralizing maternal antibody generated from maternal immunization would be expected to reduce IL-33 production and mitigate ILC2 activation. Here we report that lung ILC2s from mice born to RSV-immunized dams become activated despite undetectable RSV replication. We also report, for the first time, expression of activating and inhibitory Fcgamma receptors on ILC2s that are differentially expressed in offspring born to immunized versus unimmunized dams. Alternatively, ex vivo IL-33-mediated activation of ILC2s was mitigated following the addition of antibody: antigen immune complexes. Further studies are needed to confirm the role of Fcgamma receptor ligation by immune complexes as an alternative mechanism of ILC2 regulation in RSV-associated eosinophilic lung inflammation.


Subject(s)
Interleukin-33 , Lung , Mice, Inbred BALB C , Respiratory Syncytial Virus Infections , Respiratory Syncytial Viruses , Animals , Respiratory Syncytial Virus Infections/immunology , Respiratory Syncytial Virus Infections/prevention & control , Mice , Female , Lung/immunology , Lung/virology , Interleukin-33/immunology , Respiratory Syncytial Viruses/immunology , Lymphocytes/immunology , Immunization , Receptors, IgG/immunology , Receptors, IgG/metabolism , Antibodies, Viral/immunology , Pregnancy , Respiratory Syncytial Virus Vaccines/immunology
2.
Cancer Lett ; 575: 216396, 2023 Oct 28.
Article in English | MEDLINE | ID: mdl-37739209

ABSTRACT

Recent discoveries in cancer metabolism have revealed promising metabolic targets to modulate cancer progression, drug response, and anti-cancer immunity. Combination therapy, consisting of metabolic inhibitors and chemotherapeutic or immunotherapeutic agents, offers new opportunities for improved cancer therapy. However, it also presents challenges due to the complexity of cancer metabolic pathways and the metabolic interactions between tumor cells and immune cells. Many studies have been published demonstrating potential synergy between novel inhibitors of metabolism and chemo/immunotherapy, yet our understanding of the underlying mechanisms remains limited. Here, we review the current strategies of altering the metabolic pathways of cancer to improve the anti-cancer effects of chemo/immunotherapy. We also note the need to differentiate the effect of metabolic inhibition on cancer cells and immune cells and highlight nanotechnology as an emerging solution. Improving our understanding of the complexity of the metabolic pathways in different cell populations and the anti-cancer effects of chemo/immunotherapy will aid in the discovery of novel strategies that effectively restrict cancer growth and augment the anti-cancer effects of chemo/immunotherapy.

3.
Front Immunol ; 14: 1206026, 2023.
Article in English | MEDLINE | ID: mdl-37646035

ABSTRACT

Respiratory syncytial virus (RSV) is the leading cause of childhood hospitalizations due to bronchiolitis in children under 5 years of age. Moreover, severe RSV disease requiring hospitalization is associated with the subsequent development of wheezing and asthma. Due to the young age in which viral protection is needed and risk of vaccine enhanced disease following direct infant vaccination, current approaches aim to protect young children through maternal immunization strategies that boost neutralizing maternal antibody (matAb) levels. However, there is a scarcity of studies investigating the influence of maternal immunization on secondary immune responses to RSV in the offspring or whether the subsequent development of wheezing and asthma is mitigated. Toward this goal, our lab developed a murine model of maternal RSV vaccination and repeat RSV exposure to evaluate the changes in immune response and development of exacerbated lung inflammation on secondary RSV exposure in mice born to immunized dams. Despite complete protection following primary RSV exposure, offspring born to pre-fusion F (PreF)-vaccinated dams had exaggerated secondary ILC2 and Th2 responses, characterized by enhanced production of IL-4, IL-5, and IL-13. These enhanced type 2 cellular responses were associated with exaggerated airway eosinophilia and mucus hyperproduction upon re-exposure to RSV. Importantly, depletion of CD4+ T cells led to complete amelioration of the observed type 2 pathology on secondary RSV exposure. These unanticipated results highlight the need for additional studies that look beyond primary protection to better understand how maternal immunization shapes subsequent immune responses to repeat RSV exposure.


Subject(s)
Asthma , Pneumonia , Respiratory Syncytial Virus, Human , Animals , Mice , CD4-Positive T-Lymphocytes , Immunity, Innate , Respiratory Sounds , Pneumonia/prevention & control
4.
Front Immunol ; 13: 1025341, 2022.
Article in English | MEDLINE | ID: mdl-36268035

ABSTRACT

Respiratory syncytial virus (RSV) remains the most common cause of lower respiratory tract infections in children worldwide. Development of a vaccine has been hindered due the risk of enhanced respiratory disease (ERD) following natural RSV exposure and the young age (<6 months) at which children would require protection. Risk factors linked to the development of ERD include poorly neutralizing antibody, seronegative status (never been exposed to RSV), and a Th2-type immune response. Stabilization of the more antigenic prefusion F protein (PreF) has reinvigorated hope for a protective RSV vaccine that elicits potent neutralizing antibody. While anecdotal evidence suggests that children and adults previously exposed to RSV (seropositive) are not at risk for developing vaccine associated ERD, differences in host immune responses in seropositive and seronegative individuals that may protect against ERD remain unclear. It is also unclear if vaccine formulations that skew towards Th1- versus Th2-type immune responses increase pathology or provide greater protection in seropositive individuals. Therefore, the goal of this work was to compare the host immune response to a stabilized prefusion RSV antigen formulated alone or with Th1 or Th2 skewing adjuvants in seronegative and seropositive BALB/c mice. We have developed a novel BALB/c mouse model whereby mice are first infected with RSV (seropositive) and then vaccinated during pregnancy to recapitulate maternal immunization strategies. Results of these studies show that prior RSV infection mitigates vaccine-mediated skewing by Th1- and Th2-polarizing adjuvants that was observed in seronegative animals. Moreover, vaccination with PreF plus the Th1-skewing adjuvant, Advax, increased RSV F85-93-specific CD8 T cells in both seronegative and seropositive dams. These data demonstrate the importance of utilizing seropositive animals in preclinical vaccine studies to assess both the safety and efficacy of candidate RSV vaccines.


Subject(s)
Respiratory Syncytial Virus Infections , Respiratory Syncytial Virus Vaccines , Respiratory Syncytial Virus, Human , Mice , Animals , Antibodies, Viral , Lung , Antibodies, Neutralizing , Mice, Inbred BALB C , CD8-Positive T-Lymphocytes , Adjuvants, Immunologic
5.
Ther Adv Vaccines Immunother ; 9: 2515135520981516, 2021.
Article in English | MEDLINE | ID: mdl-33623860

ABSTRACT

Respiratory syncytial virus (RSV) is the leading cause of lower respiratory tract infections in children worldwide, with the most severe disease occurring in very young infants. Despite half a century of research there still are no licensed RSV vaccines. Difficulties in RSV vaccine development stem from a number of factors, including: (a) a very short time frame between birth and first RSV exposure; (b) interfering effects of maternal antibodies; and (c) differentially regulated immune responses in infants causing a marked T helper 2 (Th2) immune bias. This review seeks to provide an age-specific understanding of RSV immunity critical to the development of a successful pediatric RSV vaccine. Historical and future approaches to the prevention of infant RSV are reviewed, including passive protection using monoclonal antibodies or maternal immunization strategies versus active infant immunization using pre-fusion forms of RSV F protein antigens formulated with novel adjuvants such as Advax that avoid excess Th2 immune polarization.

6.
Vaccine ; 38(50): 7885-7891, 2020 11 25.
Article in English | MEDLINE | ID: mdl-33129608

ABSTRACT

Respiratory syncytial virus (RSV) commonly causes severe respiratory tract infections in infants, peaking between 2 and 6 months of age; an age at which direct vaccination is unlikely to be effective. Maternal immunization can deliver high levels of antibodies to newborns, providing immediate protection. Following natural infection, antibodies targeting the prefusion conformation of RSV F protein (PreF) have the greatest neutralizing capacity and thus, may provide infants with a high degree of RSV protection when acquired through maternal vaccination. However, the influence of anti-PreF maternal antibodies on infant immunity following RSV exposure has not been elucidated. To address this knowledge gap, offspring born to dams immunized with a RSV PreF vaccine formulation were challenged with RSV and their immune responses were analyzed over time. These studies demonstrated safety and efficacy for RSV-challenged, maternally-immunized offspring but high and waning maternal antibody levels were associated with differential innate and T cell immunity.


Subject(s)
Respiratory Syncytial Virus Infections , Respiratory Syncytial Virus Vaccines , Antibodies, Neutralizing , Antibodies, Viral , Humans , Immunization , Infant , Infant, Newborn , Respiratory Syncytial Virus Infections/prevention & control , T-Lymphocytes , Vaccination , Viral Fusion Proteins
7.
Vaccine ; 38(41): 6357-6362, 2020 09 22.
Article in English | MEDLINE | ID: mdl-32829976

ABSTRACT

Respiratory syncytial virus (RSV) is a leading cause of lower respiratory tract infections among infants with most infections occurring in the first year of life. Multiple RSV exposures are required for children to mount adult-like immune responses. Although adult RSV immunity is associated with less severe disease, the protection induced through natural infection is short-lived. Therefore, vaccination of RSV-experienced young children may accelerate immunity and provide long-term protection from RSV reinfection. However, the extent to which different Th-biased vaccine regimens influence pre-existing humoral and cellular immunity in RSV-experienced young children is unknown. To address this question, infant BALB/c mice were RSV-infected and subsequently immunized with the prefusion RSV F (PreF) antigen formulated with either a Th2-skewing (Alum) or Th1/Th2-balanced (Advax-SM) adjuvant. These studies show that both adjuvants boosted neutralizing antibody and protected from RSV reinfection, but Advax-SM adjuvant prevented the Th2-skewed immunity observed in RSV-experienced young mice immunized with PreF/Alum.


Subject(s)
Respiratory Syncytial Virus Infections , Respiratory Syncytial Virus Vaccines , Respiratory Syncytial Virus, Human , Animals , Antibodies, Viral , Lung , Mice , Mice, Inbred BALB C , Respiratory Syncytial Virus Infections/prevention & control
8.
Front Immunol ; 11: 1673, 2020.
Article in English | MEDLINE | ID: mdl-32849580

ABSTRACT

Respiratory syncytial virus (RSV) remains the most common cause of lower respiratory tract infections in children worldwide. Development of a vaccine has been hindered by the risk of developing enhanced respiratory disease (ERD) upon natural exposure to the virus. Generation of higher quality neutralizing antibodies with stabilized pre-fusion F protein antigens has been proposed as a strategy to prevent ERD. We sought to test whether there was evidence of ERD in naïve BALB/c mice immunized with an unadjuvanted, stabilized pre-fusion F protein, and challenged with RSV line 19. We further sought to determine the extent to which formulation with a Th2-biased (alum) or a more Th1/Th2-balanced (Advax-SM) adjuvant influenced cellular responses and lung pathology. When exposed to RSV, mice immunized with pre-fusion F protein alone (PreF) exhibited increased airway eosinophilia and mucus accumulation. This was further exacerbated by formulation of PreF with Alum (aluminum hydroxide). Conversely, formulation of PreF with a Th1/Th2-balanced adjuvant, Advax-SM, not only suppressed RSV viral replication, but also inhibited airway eosinophilia and mucus accumulation. This was associated with lower numbers of lung innate lymphocyte cells (ILC2s) and CD4+ T cells producing IL-5+ or IL-13+ and increased IFNγ+ CD4+ and CD8+ T cells, in addition to RSV F-specific CD8+ T cells. These data suggest that in the absence of preimmunity, stabilized PreF antigens may still be associated with aberrant Th2 responses that induce lung pathology in response to RSV infection, and can be prevented by formulation with more Th1/Th2-balanced adjuvants that enhance CD4+ and CD8+ IFNγ+ T cell responses. This may support the use of stabilized PreF antigens with Th1/Th2-balanced adjuvants like, Advax-SM, as safer alternatives to alum in RSV vaccine candidates.


Subject(s)
Adjuvants, Immunologic/pharmacology , Aluminum Hydroxide/pharmacology , Lung/drug effects , Respiratory Syncytial Virus Infections/prevention & control , Respiratory Syncytial Virus Vaccines/pharmacology , Respiratory Syncytial Viruses/drug effects , Th2 Cells/drug effects , Viral Fusion Proteins/pharmacology , Animals , Antibodies, Neutralizing/blood , Antibodies, Viral/blood , Cytokines/immunology , Cytokines/metabolism , Disease Models, Animal , Female , Immunity, Humoral/drug effects , Immunization , Immunogenicity, Vaccine/drug effects , Lung/immunology , Lung/pathology , Lung/virology , Mice, Inbred BALB C , Respiratory Syncytial Virus Infections/immunology , Respiratory Syncytial Virus Infections/pathology , Respiratory Syncytial Virus Infections/virology , Respiratory Syncytial Viruses/immunology , Respiratory Syncytial Viruses/pathogenicity , Th1-Th2 Balance/drug effects , Th2 Cells/immunology , Th2 Cells/metabolism , Th2 Cells/virology
9.
Pediatr Pulmonol ; 53(2): 145-153, 2018 02.
Article in English | MEDLINE | ID: mdl-29115050

ABSTRACT

OBJECTIVES: Respiratory syncytial virus (RSV) is a leading cause of lower respiratory tract infections worldwide, causing disproportionate morbidity and mortality in infants and children. Infants with stronger Th1 responses have less severe disease, yet little is known about the infant T-cell response within the air space. Thus, we tested the hypothesis that RSV infected infant mice would have quantitative and qualitative deficiencies in CD4+ and CD8+ T-cell populations isolated from the bronchoalveolar lavage when compared to adults and that local delivery of IFN-γ would increase airway CD4+ Tbet+ and CD8+ Tbet+ T-cell responses. METHODS: We compared the localization of T-cell responses in RSV-infected infant and adult mice and investigated the effects of local IFN-γ administration on infant cellular immunity. RESULTS: Adult CD8+ CD44HI and CD4+ CD44HI Tbet+ T-cells accumulated in the alveolar space whereas CD4+ CD44HI Tbet+ T-cells were evenly distributed between the infant lung tissue and airway and infant lungs contained higher frequencies of CD8+ T-cells. Delivery of IFN-γ to the infant airway failed to increase the accumulation of T-cells in the airspace and unexpectedly reduced CD4+ CD44HI Tbet+ T-cells. However, intranasal IFN-γ increased RSV F protein-specific CD8+ T-cells in the alveolar space. CONCLUSION: Together, these data suggest that quantitative and qualitative defects exist in the infant T-cell response to RSV but early, local IFN-γ exposure can increase the CD8+ RSV-specific T-cell response.


Subject(s)
Bronchoalveolar Lavage Fluid/immunology , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/metabolism , Respiratory Syncytial Virus Infections/immunology , Animals , Antiviral Agents/therapeutic use , CD4-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/drug effects , Disease Models, Animal , Hyaluronan Receptors/metabolism , Immunity, Cellular , Interferon-gamma/therapeutic use , Lung/immunology , Lung/metabolism , Mice , Mice, Inbred BALB C , Respiratory Syncytial Virus Infections/drug therapy , T-Box Domain Proteins/metabolism
10.
Data Brief ; 14: 272-277, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28861450

ABSTRACT

The data presented here are related to the research article entitled "Age predicts cytokine kinetics and innate immune cell activation following intranasal delivery of IFNγ and GM-CSF in a mouse model of RSV infection" (Eichinger et al., 2017) [1]. The cited manuscript demonstrated that the macrophage-stimulating cytokine, interferon gamma (IFNγ), but not granulocyte macrophage-colony stimulating factor (GM-CSF), effectively enhanced viral clearance in infant mice infected with respiratory syncytial virus (RSV) following intranasal delivery. This article describes the immune response and viral clearing effects of intranasal IFNγ in RSV-infected adult BALB/c mice demonstrating delayed production of endogenous IFNγ. The dataset is made publicly available to extrapolate the role of IFNγ in RSV-infected adult mice.

11.
Cytokine ; 97: 25-37, 2017 09.
Article in English | MEDLINE | ID: mdl-28558308

ABSTRACT

Respiratory syncytial virus (RSV) is the leading cause of lower respiratory tract infections in young children and is further associated with increased healthcare utilization and cost of care in the first years of life. Severe RSV disease during infancy has also been linked to the later development of allergic asthma, yet there remains no licensed RSV vaccine or effective treatment. Pre-clinical and clinical studies have shown that disease severity and development of allergic asthma are associated with differences in cytokine production. As a result, stimulation of the innate host immune response with immune potentiators is gaining attention for their prospective application in populations with limited immune responses to antigenic stimuli or against pathogens for which vaccines do not exist. Specifically, macrophage-activating cytokines such as interferon gamma (IFNγ) and granulocyte colony-stimulating factor (GM-CSF) are commercially available immune potentiators used to prevent infections in patients with chronic granulomatous disease and febrile neutropenia, respectively. Moreover, an increasing number of reports describe the protective function of IFNγ and GM-CSF as vaccine adjuvants. Although a positive correlation between cytokine production and age has previously been reported, little is known about age-dependent cytokine metabolism or immune activating responses in infant compared to adult lungs. Here we use a non-compartmental pharmacokinetic model in naïve and RSV-infected infant and adult BALB/c mice to determine the effect of age on IFNγ and GM-CSF elimination and innate cell activation following intranasal delivery.


Subject(s)
Granulocyte-Macrophage Colony-Stimulating Factor/administration & dosage , Immunity, Innate/drug effects , Interferon-gamma/administration & dosage , Respiratory Syncytial Virus Infections/immunology , Administration, Intranasal , Age Factors , Animals , Disease Models, Animal , Granulocyte-Macrophage Colony-Stimulating Factor/blood , Granulocyte-Macrophage Colony-Stimulating Factor/immunology , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacokinetics , Interferon-gamma/blood , Interferon-gamma/immunology , Interferon-gamma/pharmacokinetics , Kinetics , Lung/drug effects , Lung/immunology , Lung/virology , Macrophage Activation , Mice , Mice, Inbred BALB C , Prospective Studies , Respiratory Syncytial Virus Infections/virology , Respiratory Syncytial Virus, Human/physiology
12.
Proc Natl Acad Sci U S A ; 113(6): 1642-7, 2016 Feb 09.
Article in English | MEDLINE | ID: mdl-26729873

ABSTRACT

Clinical observations link respiratory virus infection and Pseudomonas aeruginosa colonization in chronic lung disease, including cystic fibrosis (CF) and chronic obstructive pulmonary disease. The development of P. aeruginosa into highly antibiotic-resistant biofilm communities promotes airway colonization and accounts for disease progression in patients. Although clinical studies show a strong correlation between CF patients' acquisition of chronic P. aeruginosa infections and respiratory virus infection, little is known about the mechanism by which chronic P. aeruginosa infections are initiated in the host. Using a coculture model to study the formation of bacterial biofilm formation associated with the airway epithelium, we show that respiratory viral infections and the induction of antiviral interferons promote robust secondary P. aeruginosa biofilm formation. We report that the induction of antiviral IFN signaling in response to respiratory syncytial virus (RSV) infection induces bacterial biofilm formation through a mechanism of dysregulated iron homeostasis of the airway epithelium. Moreover, increased apical release of the host iron-binding protein transferrin during RSV infection promotes P. aeruginosa biofilm development in vitro and in vivo. Thus, nutritional immunity pathways that are disrupted during respiratory viral infection create an environment that favors secondary bacterial infection and may provide previously unidentified targets to combat bacterial biofilm formation.


Subject(s)
Biofilms/growth & development , Immunity , Nutritional Physiological Phenomena , Pseudomonas aeruginosa/physiology , Respiratory Syncytial Virus Infections/pathology , Respiratory Syncytial Viruses/physiology , Animals , Antiviral Agents/pharmacology , Bronchi/pathology , Bronchoalveolar Lavage Fluid , Cystic Fibrosis/microbiology , Cystic Fibrosis/pathology , Epithelial Cells/drug effects , Epithelial Cells/microbiology , Epithelial Cells/virology , Homeostasis/drug effects , Humans , Interferon-beta/pharmacology , Iron/pharmacology , Mice , Microbial Interactions/drug effects , Models, Biological , Pseudomonas aeruginosa/drug effects , Respiratory Syncytial Viruses/drug effects , Signal Transduction/drug effects , Transferrin/metabolism
13.
Respir Res ; 16: 122, 2015 Oct 05.
Article in English | MEDLINE | ID: mdl-26438053

ABSTRACT

BACKGROUND: Poor interferon gamma (IFNγ) production during respiratory syncytial virus (RSV) is associated with prolonged viral clearance and increased disease severity in neonatal mice and humans. We previously showed that intra-nasal delivery of IFNγ significantly enhances RSV clearance from neonatal lungs prior to observed T-lymphocyte recruitment or activation, suggesting an innate immune mechanism of viral clearance. We further showed that alveolar macrophages dominate the RSV-infected neonatal airways relative to adults, consistent with human neonatal autopsy data. Therefore, the goal of this work was to determine the role of neonatal alveolar macrophages in IFNγ-mediated RSV clearance. METHODS: Clodronate liposomes, flow cytometry, viral plaque assays, and histology were used to examine the role of alveolar macrophages (AMs) and the effects of intra-nasal IFNγ in RSV infected neonatal Balb/c mice. The functional outcomes of AM depletion were determined quantitatively by viral titers using plaque assay. Illness was assessed by measuring reduced weight gain. RESULTS: AM activation during RSV infection was age-dependent and correlated tightly with IFNγ exposure. Higher doses of IFNγ more efficiently stimulated AM activation and expedited RSV clearance without significantly affecting weight gain. The presence of AMs were independently associated with improved RSV clearance, whereas AM depletion but not IFNγ exposure, significantly impaired weight gain in RSV-infected neonates. CONCLUSION: We show here for the first time, that IFNγ is critical for neonatal RSV clearance and that it depends, in part, on alveolar macrophages (AMs) for efficient viral clearing effects. Early reductions in viral burden are likely to have profound short- and long-term immune effects in the vulnerable post-natally developing lung environment. Studies are ongoing to elucidate the pathologic effects associated with early versus delayed RSV clearance in developing neonatal airways.


Subject(s)
Antiviral Agents/administration & dosage , Interferon-gamma/administration & dosage , Macrophage Activation/drug effects , Macrophages, Alveolar/drug effects , Respiratory Syncytial Virus Infections/drug therapy , Respiratory Syncytial Viruses/drug effects , Administration, Intranasal , Age Factors , Animals , Animals, Newborn , Antiviral Agents/pharmacokinetics , Dendritic Cells/drug effects , Dendritic Cells/immunology , Disease Models, Animal , Dose-Response Relationship, Drug , Female , Interferon-gamma/pharmacokinetics , Macrophages, Alveolar/immunology , Macrophages, Alveolar/metabolism , Macrophages, Alveolar/virology , Male , Mice, Inbred BALB C , Mice, Knockout , Receptors, Interferon/deficiency , Receptors, Interferon/genetics , Respiratory Syncytial Virus Infections/immunology , Respiratory Syncytial Virus Infections/metabolism , Respiratory Syncytial Virus Infections/virology , Respiratory Syncytial Viruses/immunology , Respiratory Syncytial Viruses/pathogenicity , Time Factors , Viral Load , Weight Gain , Interferon gamma Receptor
14.
Ann Pharmacother ; 48(1): 77-85, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24259637

ABSTRACT

OBJECTIVE: To evaluate the evidence describing the immunosuppressive and pharmacokinetic properties of commonly used analgesic and sedation agents in critically ill patients. DATA SOURCES: MEDLINE (January 1980-September 2013) was searched. STUDY SELECTION AND DATA EXTRACTION: All in vitro and in vivo studies that evaluated the immune-modulating properties of analgesic and sedation agents commonly used in the critically ill were included. Full-text and abstract-only articles (noted) were included in this review. Inclusion criteria were met by 46 studies and were evaluated. DATA SYNTHESIS: Analgesic and sedation agents have been shown to be immunosuppressive in a variety of models. In vitro models use a variety of immune cells to demonstrate the immunosuppressive properties of opioids, benzodiazepines, and to a lesser extent, propofol. In each case, animal studies provide more robust data supporting the concept that opioids, benzodiazepines, and propofol exhibit immunosuppressive activities ranging from innate to adaptive immune alterations. Human studies, though more limited, provide further support that these agents inhibit the immune response. In contrast, data have shown that dexmedetomidine may attenuate the immune system. Clinical trial data evaluating the immunosuppressive properties of these agents is limited. CONCLUSIONS: Analgesic and sedation agents have clearly been shown to alter cellular function and other mediators of the immune system; yet the clinical impact remains to be fully elucidated. The mechanism by which sedation interruption reduces ventilator-associated pneumonia may in fact be a reduction in immunosuppressive effects. Studies linking the immune-modulating effects of analgesic and sedation agents in critically ill patients are needed.


Subject(s)
Analgesics/adverse effects , Hypnotics and Sedatives/adverse effects , Immunosuppressive Agents/adverse effects , Pneumonia, Ventilator-Associated/epidemiology , Respiration, Artificial/adverse effects , Animals , Humans , Pneumonia, Ventilator-Associated/etiology , Risk Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...