Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Wound Repair Regen ; 27(6): 622-633, 2019 11.
Article in English | MEDLINE | ID: mdl-31276609

ABSTRACT

Literature describes that a well-maintained moist wound healing environment leads to faster healing by preventing scabbing and drying of the wound. A moist wound speeds healing by allowing for unimpeded movement of newly dividing epidermal cells in the wound. Contrary to what is described in literature and practiced by clinicians, first-aid dressings used at home by consumers advertise breathability and absorptivity as benefits. This manuscript examines the effects of dressing breathability and highly absorptive pads on healing and wound appearance in a porcine dermatome wound model, designed to mimic an abrasion injury. Partial thickness wounds were covered with an experimental silicone-polymer film dressing and various over-the-counter bandages for time frames ranging from 4 to 11 days. The progression of healing was quantified by histology and wound-size reduction measurements. The thickness and persistence of a scab or serocellular crust (SCC) over the injury was measured using both pixel density and optical coherence tomography to supplement visual observations, demonstrating new tools for quantification of SCC over wounds. The results of the experiments illustrate the impact of dressing features on the rate of wound reepithelialization and the formation of SCC. Both a low moisture vapor transmission rate (MVTR) and the absence of an absorptive layer were important in speeding wound healing. Surprisingly, use of a dressing with a low MVTR and a highly absorptive pad healed significantly more slowly than a comparative dressing with a low MVTR and no absorptive pad, even though both dressings had very little scab formation over the wound. This study shows that breathability and absorbency of dressings play independent roles in providing an optimal healing environment, and that these properties can vary widely among commercially available dressings.


Subject(s)
Bandages , First Aid/methods , Skin/injuries , Wound Healing/physiology , Wounds and Injuries/pathology , Wounds and Injuries/therapy , Animals , Biopsy, Needle , Disease Models, Animal , Female , Immunohistochemistry , Injury Severity Score , Re-Epithelialization/physiology , Silicones/pharmacology , Skin/pathology , Swine , Time Factors
2.
J Allergy Clin Immunol ; 140(5): 1339-1350, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28343701

ABSTRACT

BACKGROUND: Newborns display distinct immune responses, leaving them vulnerable to infections and impairing immunization. Targeting newborn dendritic cells (DCs), which integrate vaccine signals into adaptive immune responses, might enable development of age-specific vaccine formulations to overcome suboptimal immunization. OBJECTIVE: Small-molecule imidazoquinoline Toll-like receptor (TLR) 8 agonists robustly activate newborn DCs but can result in reactogenicity when delivered in soluble form. We used rational engineering and age- and species-specific modeling to construct and characterize polymer nanocarriers encapsulating a TLR8 agonist, allowing direct intracellular release after selective uptake by DCs. METHODS: Chemically similar but morphologically distinct nanocarriers comprised of amphiphilic block copolymers were engineered for targeted uptake by murine DCs in vivo, and a range of TLR8 agonist-encapsulating polymersome formulations were then synthesized. Novel 96-well in vitro assays using neonatal human monocyte-derived DCs and humanized TLR8 mouse bone marrow-derived DCs enabled benchmarking of the TLR8 agonist-encapsulating polymersome formulations against conventional adjuvants and licensed vaccines, including live attenuated BCG vaccine. Immunogenicity of the TLR8 agonist adjuvanted antigen 85B (Ag85B)/peptide 25-loaded BCG-mimicking nanoparticle formulation was evaluated in vivo by using humanized TLR8 neonatal mice. RESULTS: Although alum-adjuvanted vaccines induced modest costimulatory molecule expression, limited TH-polarizing cytokine production, and significant cell death, BCG induced a robust adult-like maturation profile of neonatal DCs. Remarkably, TLR8 agonist polymersomes induced not only newborn DC maturation profiles similar to those induced by BCG but also stronger IL-12p70 production. On subcutaneous injection to neonatal mice, the TLR8 agonist-adjuvanted Ag85B peptide 25 formulation was comparable with BCG in inducing Ag85B-specific CD4+ T-cell numbers. CONCLUSION: TLR8 agonist-encapsulating polymersomes hold substantial potential for early-life immunization against intracellular pathogens. Overall, our study represents a novel approach for rational design of early-life vaccines.


Subject(s)
Adjuvants, Immunologic/administration & dosage , BCG Vaccine/immunology , Dendritic Cells/immunology , Imidazoles/administration & dosage , Monocytes/immunology , Nanoparticles/administration & dosage , Quinolines/administration & dosage , Adaptive Immunity , Animals , Animals, Newborn , Biomimetics , CD4-Positive T-Lymphocytes/immunology , Cells, Cultured , Cytokines/metabolism , Humans , Imidazoles/chemistry , Imidazoles/pharmacology , Immunity, Innate , Immunomodulation , Infant, Newborn , Mice , Mice, Inbred C57BL , Mice, SCID , Nanoparticles/chemistry , Polymers/chemistry , Quinolines/chemistry , Quinolines/pharmacology , Toll-Like Receptor 8/agonists , Vaccination
4.
Sci Rep ; 6: 21094, 2016 Feb 19.
Article in English | MEDLINE | ID: mdl-26891899

ABSTRACT

Opioid drug abusers have a greater susceptibility to gram positive (Gram (+)) bacterial infections. However, the mechanism underlying opioid modulation of Gram (+) versus Gram (-) bacterial clearance has not been investigated. In this study, we show that opioid treatment resulted in reduced phagocytosis of Gram (+), when compared to Gram (-) bacteria. We further established that LPS priming of chronic morphine treated macrophages leads to potentiated phagocytosis and killing of both Gram (+) and Gram (-) bacteria in a P-38 MAP kinase dependent signaling pathway. In contrast, LTA priming lead to inhibition of both phagocytosis and bacterial killing. This study demonstrates for the first time the differential effects of TLR4 and TLR2 agonists on morphine induced inhibition of phagocytosis. Our results suggest that the incidence and severity of secondary infections with Gram (+) bacteria would be higher in opioid abusers.


Subject(s)
Gram-Negative Bacteria/metabolism , Gram-Positive Bacteria/metabolism , Lipopolysaccharides/pharmacology , Morphine/pharmacology , Phagocytosis/drug effects , Animals , Gene Expression , Gram-Negative Bacteria/immunology , Gram-Positive Bacteria/immunology , Immunomodulation , Ligands , Lipopolysaccharides/immunology , Macrophages/drug effects , Macrophages/immunology , Macrophages/metabolism , Mice , Mice, Knockout , Microbial Viability/drug effects , Microbial Viability/immunology , Models, Biological , Phagocytosis/immunology , Teichoic Acids/immunology , Teichoic Acids/pharmacology , Toll-Like Receptor 2/genetics , Toll-Like Receptor 2/metabolism , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/metabolism
5.
Sci Rep ; 5: 11384, 2015 Jun 15.
Article in English | MEDLINE | ID: mdl-26072707

ABSTRACT

Opportunistic lung infection and inflammation is a hallmark of chronic recreational/clinical use of morphine. We show that early induction of IL17 from the bronchial epithelium, following pathogenic encounter is a protective response, which contributes to pathogenic clearance and currently attributed to TLR2 activation in immune cells. Concurrent activation of TLR2 and IL17R in bronchial epithelium results in the sequestration of MyD88 (TLR2 adapter) by Act1/CIKS (IL17R adapter), thereby turning off TLR2 signaling to restore homeostasis. Morphine inhibits the early IL17 release and interaction between Act1 and MyD88, leading to decreased pathogenic clearance and sustained inflammation. Hence, we propose that therapeutically targeting either TLR2 or IL17 in bronchial epithelia, in the context of morphine, can restore inflammatory homeostasis.


Subject(s)
Interleukin-17/immunology , Lung/immunology , Morphine/adverse effects , Pneumococcal Infections/immunology , Receptors, Interleukin-17/immunology , Toll-Like Receptor 2/immunology , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/immunology , Animals , Gene Expression Regulation , Homeostasis , Humans , Inflammation , Interleukin-17/genetics , Lung/microbiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Myeloid Differentiation Factor 88/genetics , Myeloid Differentiation Factor 88/immunology , Pneumococcal Infections/etiology , Pneumococcal Infections/microbiology , Pneumococcal Infections/pathology , Receptor Cross-Talk/drug effects , Receptor Cross-Talk/immunology , Receptors, Interleukin-17/genetics , Receptors, Opioid, mu/deficiency , Receptors, Opioid, mu/genetics , Receptors, Opioid, mu/immunology , Signal Transduction , Streptococcus pneumoniae/immunology , Toll-Like Receptor 2/genetics , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/immunology
6.
J Infect Dis ; 212(9): 1480-90, 2015 Nov 01.
Article in English | MEDLINE | ID: mdl-25883383

ABSTRACT

BACKGROUND: Staphylococcus epidermidis causes late-onset sepsis in preterm infants. Staphylococcus epidermidis activates host responses in part via Toll-like receptor 2 (TLR2). Epidemiologic studies link bacteremia and neonatal brain injury, but direct evidence is lacking. METHODS: Wild-type and TLR2-deficient (TLR2-/-) mice were injected intravenously with S. epidermidis at postnatal day 1 prior to measuring plasma and brain cytokine and chemokine levels, bacterial clearance, brain caspase-3 activation, white/gray matter volume, and innate transcriptome. RESULTS: Staphylococcus epidermidis bacteremia spontaneously resolved over 24 hours without detectable bacteria in the cerebrospinal fluid (CSF). TLR2-/- mice demonstrated delayed S. epidermidis clearance from blood, spleen, and liver. Staphylococcus epidermidis increased the white blood cell count in the CSF, increased interleukin 6, interleukin 12p40, CCL2, and CXCL1 concentrations in plasma; increased the CCL2 concentration in the brain; and caused rapid (within 6 hours) TLR2-dependent brain activation of caspase-3 and TLR2-independent white matter injury. CONCLUSIONS: Staphylococcus epidermidis bacteremia, in the absence of bacterial entry into the CSF, impairs neonatal brain development. Staphylococcus epidermidis bacteremia induced both TLR2-dependent and -independent brain injury, with the latter occurring in the absence of TLR2, a condition associated with an increased bacterial burden. Our study indicates that the consequences of transient bacteremia in early life may be more severe than commonly appreciated, and our findings may inform novel approaches to reduce bacteremia-associated brain injury.


Subject(s)
Bacteremia/pathology , Brain Injuries/microbiology , Staphylococcus epidermidis/isolation & purification , Toll-Like Receptor 2/metabolism , Animals , Animals, Newborn , Caspase 3/genetics , Caspase 3/metabolism , Chemokine CCL2/blood , Chemokine CXCL1/blood , Colony Count, Microbial , Disease Models, Animal , Interleukin-12 Subunit p40/blood , Interleukin-6/blood , Liver/microbiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Spleen/microbiology , Toll-Like Receptor 2/genetics , Up-Regulation
7.
J Vis Exp ; (93): e52195, 2014 Nov 27.
Article in English | MEDLINE | ID: mdl-25489737

ABSTRACT

The goal of fluorometric analysis is to serve as an efficient, cost effective, high throughput method of analyzing phagocytosis and other cellular processes. This technique can be used on a variety of cell types, both adherent and non-adherent, to examine a variety of cellular properties. When studying phagocytosis, fluorometric technique utilizes phagocytic cell types such as macrophages, and fluorescently labeled opsonized particles whose fluorescence can be extinguished in the presence of trypan blue. Following plating of adherent macrophages in 96-well plates, fluorescent particles (green or red) are administered and cells are allowed to phagocytose for varied amounts of time. Following internalization of fluorescent particles, cells are washed with trypan blue, which facilitates extinction of fluorescent signal from bacteria which are not internalized, or are merely adhering to the cell surface. Following the trypan wash, cells are washed with PBS, fixed, and stained with DAPI (nuclear blue fluorescent label), which serves to label nuclei of cells. By a simple fluorometric quantification through plate reading of nuclear (blue) or particle (red/green) fluorescence we can examine the ratio of relative fluorescence units of green:blue and determine a phagocytic index indicative of amount of fluorescent bacteria internalized per cell. The duration of assay using a 96-well method and multichannel pipettes for washing, from end of phagocytosis to end of data acquisition, is less than 45 min. Flow cytometry could be used in a similar manner but the advantage of fluorometry is its high throughput, rapid method of assessment with minimal manipulation of samples and quick quantification of fluorescent intensity per cell. Similar strategies can be applied to non adherent cells, live labeled bacteria, actin polymerization, and essentially any process utilizing fluorescence. Therefore, fluorometry is a promising method for its low cost, high throughput capabilities in the study of cellular processes.


Subject(s)
Actins/metabolism , Fluorometry/methods , Macrophages/chemistry , Animals , Fluorescent Dyes/chemistry , Humans , Macrophages/immunology , Macrophages/metabolism , Mice , Phagocytosis/immunology , Polymerization , Trypan Blue/chemistry
8.
Biomaterials ; 35(31): 8876-8886, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25023392

ABSTRACT

Neonates and infants are susceptible to infection due to distinct immune responses in early life. Therefore, development of vaccine formulation and delivery systems capable of activating human newborn leukocytes is of global health importance. Poly[di(carboxylatophenoxy)phosphazene] (PCPP) belongs to a family of ionic synthetic polyphosphazene polyelectrolyte compounds that can form non-covalent interactions with protein antigens and demonstrate adjuvant activity in animals and in human clinical trials. However, little is known about their ability to activate human immune cells. In this study, we characterized the effects of PCPP alone or in combination with a model antigen (recombinant HIV-Gag (Gag)), on the maturation, activation and antigen presentation by human adult and newborn dendritic cells (DCs) in vitro. PCPP treatment induced DC activation as assessed by upregulation of co-stimulatory molecules and cytokine production. Studies benchmarking PCPP to Alum, the most commonly used vaccine adjuvant, demonstrated that both triggered cell death and release of danger signals in adult and newborn DCs. When complexed with Gag antigen, PCPP maintained its immunostimulatory characteristics while permitting internalization and presentation of Gag by DCs to HIV-Gag-specific CD4(+) T cell clones. The PCPP vaccine formulation outlined here has intrinsic adjuvant activity, can facilitate effective delivery of antigen to DCs, and may be advantageous for induction of beneficial T cell-mediated immunity. Moreover, polyphosphazenes can further reduce cost of vaccine production and distribution through their dose-sparing and antigen-stabilizing properties, thus potentially eliminating the need for cold chain distribution.


Subject(s)
Adjuvants, Immunologic/pharmacology , Dendritic Cells/drug effects , Organophosphorus Compounds/pharmacology , Polymers/pharmacology , gag Gene Products, Human Immunodeficiency Virus/pharmacology , Adult , Alum Compounds/pharmacology , Antigen Presentation/drug effects , Cells, Cultured , Dendritic Cells/immunology , Humans , Immunity, Cellular/drug effects , Infant , Recombinant Proteins/pharmacology
9.
Amino Acids ; 45(1): 9-24, 2013 Jul.
Article in English | MEDLINE | ID: mdl-22170499

ABSTRACT

Endogenous opioids are synthesized in vivo to modulate pain mechanisms and inflammatory pathways. Endogenous and exogenous opioids mediate analgesia in response to painful stimuli by binding to opioid receptors on neuronal cells. However, wide distribution of opioid receptors on tissues and organ systems outside the CNS, such as the cells of the immune system, indicate that opioids are capable of exerting additional effects in the periphery, such as immunomodulation. The increased prevalence of infections in opioid abuser-based epidemiological studies further highlights the immunosuppressive effects of opioids. In spite of their many debilitating side effects, prescription opioids remain a gold standard for treatment of chronic pain. Therefore, given the prevalence of opioid use and abuse, opioid-mediated immune suppression presents a serious concern in our society today. It is imperative to understand the mechanisms by which exogenous opioids modulate immune processes. In this review, we will discuss the role of opioid receptors and their ligands in mediating immune-suppressive functions. We will summarize recent studies on direct and indirect opioid modulation of the cells of the immune system, as well as the role of opioids in exacerbation of certain disease states.


Subject(s)
Analgesics, Opioid/pharmacology , Immunity, Cellular/drug effects , Immunity, Innate/drug effects , Opioid Peptides/metabolism , Receptors, Opioid, mu/metabolism , Animals , Binding Sites , Humans , Immunomodulation , Leukocytes, Mononuclear/metabolism , Morphine/pharmacology , Pain/metabolism , Signal Transduction
10.
PLoS One ; 7(9): e43897, 2012.
Article in English | MEDLINE | ID: mdl-22970147

ABSTRACT

Staphylococcus epidermidis (SE) causes late onset sepsis and significant morbidity in catheterized preterm newborns. Animal models of SE infection are useful in characterizing disease mechanisms and are an important approach to developing improved diagnostics and therapeutics. Current murine models of neonatal bacterial infection employ intraperitoneal or subcutaneous routes at several days of age, and may, therefore, not accurately reflect distinct features of innate immune responses to bacteremia. In this study we developed, validated, and characterized a murine model of intravenous (IV) infection in neonatal mice <24 hours (h) old to describe the early innate immune response to SE. C57BL/6 mice <24 h old were injected IV with 10(6), 10(7), 10(8) colony-forming units (CFU) of SE 1457, a clinical isolate from a central catheter infection. A prospective injection scoring system was developed and validated, with only high quality injections analyzed. Newborn mice were euthanized between 2 and 48 h post-injection and spleen, liver, and blood collected to assess bacterial viability, gene expression, and cytokine production. High quality IV injections demonstrated inoculum-dependent infection of spleen, liver and blood. Within 2 h of injection, SE induced selective transcription of TLR2 and MyD88 in the liver, and increased systemic production of plasma IL-6 and TNF-α. Despite clearance of bacteremia and solid organ infection within 48 h, inoculum-dependent impairment in weight gain was noted. We conclude that a model of IV SE infection in neonatal mice <24 h old is feasible, demonstrating inoculum-dependent infection of solid organs and a pattern of bacteremia, rapid and selective innate immune activation, and impairment of weight gain typical of infected human neonates. This novel model can now be used to characterize immune ontogeny, evaluate infection biomarkers, and assess preventative and therapeutic modalities.


Subject(s)
Immunity, Innate/immunology , Staphylococcal Infections/immunology , Staphylococcal Infections/microbiology , Staphylococcus epidermidis/immunology , Animals , Animals, Newborn , Bacteremia/blood , Bacteremia/genetics , Bacteremia/immunology , Disease Models, Animal , Feasibility Studies , Gene Expression Regulation , Humans , Immunity, Innate/genetics , Injections, Intravenous , Interleukin-6/biosynthesis , Liver/metabolism , Liver/microbiology , Liver/pathology , Mice , Mice, Inbred C57BL , Organ Specificity/genetics , RNA, Messenger/genetics , RNA, Messenger/isolation & purification , Signal Transduction/genetics , Signal Transduction/immunology , Staphylococcal Infections/blood , Staphylococcal Infections/genetics , Toll-Like Receptors/genetics , Toll-Like Receptors/metabolism , Transcription, Genetic , Transcriptome/genetics , Tumor Necrosis Factor-alpha/biosynthesis , Weight Gain/genetics , Weight Gain/immunology
11.
Am J Pathol ; 180(3): 1068-1079, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22248582

ABSTRACT

Morphine increases the susceptibility to opportunistic infection by attenuating bacterial clearance through inhibition of Fcγ receptor (FcgR)-mediated phagocytosis. Mechanisms by which morphine inhibits this process remain to be investigated. Actin polymerization is essential for FcgR-mediated internalization; therefore, disruption of the signaling mechanisms involved in this process is detrimental to the phagocytic ability of macrophages. To our knowledge, this study is the first to propose the modulation of actin polymerization and upstream signaling effectors [cAMP, Rac1-GTP, and p38 mitogen-activated protein kinase (MAPK)] as key mechanisms by which morphine leads to inhibition of pathogen clearance. Our results indicate that long-term morphine treatment in vitro and in vivo, through activation of the µ-opioid receptor, leads to an increase in intracellular cAMP, activation of protein kinase A, and inhibition of Rac1-GTPase and p38 MAPK, thereby attenuating actin polymerization and reducing membrane ruffling. Furthermore, because of long-term morphine treatment, FcgR-mediated internalization of opsonized dextran beads is also reduced. Morphine's inhibition of Rac1-GTPase activation is abolished in J774 macrophages transfected with constitutively active pcDNA3-EGFP-Rac1-Q61L plasmid. Dibutyryl-cAMP inhibits, whereas H89 restores, activation of Rac-GTPase and abolishes morphine's inhibitory effect, implicating cAMP as the key effector in morphine's modulation of actin polymerization. These findings indicate that long-term morphine treatment, by increasing intracellular cAMP and activating protein kinase A, leads to inhibition of Rac1-GTPase and p38 MAPK, causing attenuation of actin polymerization, FcgR-mediated phagocytosis, and decreased bacterial clearance.


Subject(s)
Actins/metabolism , Cyclic AMP/metabolism , Morphine/pharmacology , Phagocytosis/drug effects , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , rac1 GTP-Binding Protein/antagonists & inhibitors , Animals , Cyclic AMP-Dependent Protein Kinases/metabolism , Isoquinolines/pharmacology , MAP Kinase Signaling System/physiology , Macrophages, Peritoneal/drug effects , Macrophages, Peritoneal/physiology , Mice , Mice, Knockout , Polymerization , Protein Kinase Inhibitors/pharmacology , Receptors, IgG/metabolism , Receptors, Opioid, mu/antagonists & inhibitors , Sulfonamides/pharmacology , Time Factors
12.
J Neuroimmune Pharmacol ; 6(4): 442-65, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21789507

ABSTRACT

Infection rate among intravenous drug users (IDU) is higher than the general public, and is the major cause of morbidity and hospitalization in the IDU population. Epidemiologic studies provide data on increased prevalence of opportunistic bacterial infections such as TB and pneumonia, and viral infections such as HIV-1 and hepatitis in the IDU population. An important component in the intravenous drug abuse population and in patients receiving medically indicated chronic opioid treatment is opioid withdrawal. Data on bacterial virulence in the context of opioid withdrawal suggest that mice undergoing withdrawal had shortened survival and increased bacterial load in response to Salmonella infection. As the body of evidence in support of opioid dependency and its immunosuppressive effects is growing, it is imperative to understand the mechanisms by which opioids exert these effects and identify the populations at risk that would benefit the most from the interventions to counteract opioid immunosuppressive effects. Thus, it is important to refine the existing animal model to closely match human conditions and to cross-validate these findings through carefully controlled human studies. Better understanding of the mechanisms will facilitate the search for new therapeutic modalities to counteract adverse effects including increased infection rates. This review will summarize the effects of morphine on innate and adaptive immunity, identify the role of the mu opioid receptor in these functions and the signal transduction activated in the process. The role of opioid withdrawal in immunosuppression and the clinical relevance of these findings will also be discussed.


Subject(s)
Analgesics, Opioid/immunology , Immune System/drug effects , Opportunistic Infections/chemically induced , Opportunistic Infections/immunology , Substance-Related Disorders/immunology , Analgesics, Opioid/pharmacology , Animals , Humans , Morphine/pharmacology , Morphine Dependence/immunology , Morphine Dependence/microbiology , Opportunistic Infections/metabolism , Receptors, Opioid, mu/metabolism , Substance Withdrawal Syndrome/immunology , Substance Withdrawal Syndrome/metabolism , Substance-Related Disorders/metabolism
13.
J Immunol ; 180(6): 3670-9, 2008 Mar 15.
Article in English | MEDLINE | ID: mdl-18322172

ABSTRACT

There are very few studies that examine the effects that morphine withdrawal has on immune functioning, and of these even fewer describe the mechanisms by which withdrawal brings about these changes. Our previous work demonstrated that morphine withdrawal contributed to Th cell differentiation by biasing cells toward the Th2 lineage. A major finding from these studies was that IL-12 was decreased following withdrawal, and it was concluded that this decrease may be a mechanism by which morphine withdrawal is mediating Th2 polarization. Therefore, it was the aim of the current studies to develop an in vitro model to examine the process of morphine withdrawal and to understand the signaling mechanisms that withdrawal may use to effect IL-12 production through the use of this model. It was demonstrated and concluded that morphine withdrawal may be effecting IL-12 production by increasing cAMP levels, which activates protein kinase A. Protein kinase A activation then prevents the phosphorylation and subsequent degradation of IkappaB, which in turn prevents translocation of the NF-kappaB p65 subunit to the nucleus to transactivate the IL-12 p40 gene, ultimately resulting in decreased IL-12 production following LPS stimulation.


Subject(s)
Cyclic AMP/physiology , Down-Regulation/immunology , Immunosuppressive Agents/toxicity , Interleukin-12/antagonists & inhibitors , Interleukin-12/biosynthesis , Macrophages, Alveolar/immunology , Morphine/toxicity , Substance Withdrawal Syndrome/immunology , Animals , Cell Line , Cells, Cultured , Disease Models, Animal , Down-Regulation/drug effects , Humans , Immunosuppressive Agents/administration & dosage , Interleukin-12 Subunit p40/antagonists & inhibitors , Interleukin-12 Subunit p40/biosynthesis , Lipopolysaccharides/pharmacology , Macrophages, Alveolar/drug effects , Macrophages, Alveolar/metabolism , Male , Mice , Mice, Inbred C57BL , Morphine/administration & dosage , Signal Transduction/drug effects , Signal Transduction/immunology , Spleen/cytology , Spleen/immunology , Spleen/metabolism , Substance Withdrawal Syndrome/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...