Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 241
Filter
1.
bioRxiv ; 2023 Dec 14.
Article in English | MEDLINE | ID: mdl-38168302

ABSTRACT

Background: The inability to evaluate host immunity in a rapid quantitative manner in patients with sepsis has severely hampered development of novel immune therapies. The ELISpot assay is a functional bioassay that measures the number of cytokine-secreting cells and the relative amount of cytokine produced at the single-cell level. A key advantage of ELISpot is its excellent dynamic range enabling a more precise quantifiable assessment of host immunity. Herein, we tested the hypothesis on whether the ELISpot assay can detect dynamic changes in both innate and adaptive immunity as they often occur during sepsis. We also tested whether ELISpot could detect the effect of immune drug therapies to modulate innate and adaptive immunity. Methods: Mice were made septic using sublethal cecal ligation and puncture (CLP). Blood and spleens were harvested serially and ex vivo IFN-γ and TNF-α production were compared by ELISpot and ELISA. The capability of ELISpot to detect changes in innate and adaptive immunity due to in vivo immune therapy with dexamethasone, IL-7, and arginine was also evaluated. Results: ELISpot confirmed a decreased innate and adaptive immunity responsiveness during sepsis progression. More importantly, ELISpot was also able to detect changes in adaptive and innate immunity in response to immune-modulatory reagents, for example dexamethasone, arginine, and IL-7 in a readily quantifiable manner, as predicted by the reagents known mechanisms of action. ELISpot and ELISA results tended to parallel one another although some differences were noted. Conclusion: ELISpot offers a unique capability to assess the functional status of both adaptive and innate immunity over time. The results presented herein demonstrate that ELISpot can also be used to detect and follow the in vivo effects of drugs to ameliorate sepsis-induced immune dysfunction. This capability would be a major advance in guiding new immune therapies in sepsis.

2.
Clin Exp Immunol ; 191(3): 268-278, 2018 03.
Article in English | MEDLINE | ID: mdl-29052227

ABSTRACT

The high mortality in neonatal sepsis has been related to both quantitative and qualitative differences in host protective immunity. Pretreatment strategies to prevent sepsis have received inadequate consideration, especially in the premature neonate, where outcomes from sepsis are so dismal. Aluminium salts-based adjuvants (alum) are used currently in many paediatric vaccines, but their use as an innate immune stimulant alone has not been well studied. We asked whether pretreatment with alum adjuvant alone could improve outcome and host innate immunity in neonatal mice given polymicrobial sepsis. Subcutaneous alum pretreatment improves survival to polymicrobial sepsis in both wild-type and T and B cell-deficient neonatal mice, but not in caspase-1/11 null mice. Moreover, alum increases peritoneal macrophage and neutrophil phagocytosis, and decreases bacterial colonization in the peritoneum. Bone marrow-derived neutrophils from alum-pretreated neonates produce more neutrophil extracellular traps (NETs) and exhibit increased expression of neutrophil elastase (NE) after in-vitro stimulation with phorbol esters. In addition, alum pretreatment increases bone marrow and splenic haematopoietic stem cell expansion following sepsis. Pretreatment of neonatal mice with an alum-based adjuvant can stimulate multiple innate immune cell functions and improve survival. These novel findings suggest a therapeutic pathway for the use of existing alum-based adjuvants for preventing sepsis in premature infants.


Subject(s)
Adjuvants, Immunologic , Alum Compounds/therapeutic use , Bacterial Vaccines/immunology , Macrophages, Peritoneal/immunology , Myeloid Cells/physiology , Neutrophils/immunology , Sepsis/immunology , Animals , Animals, Newborn , B-Lymphocytes/physiology , Caspase 1/genetics , Caspase 1/metabolism , Caspases/genetics , Caspases/metabolism , Caspases, Initiator , Cell Self Renewal , Disease Models, Animal , Extracellular Traps/metabolism , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Phagocytosis , Sepsis/prevention & control , T-Lymphocytes/physiology
3.
Eur J Trauma Emerg Surg ; 37(6): 549-58, 2011 Dec.
Article in English | MEDLINE | ID: mdl-26815465

ABSTRACT

Despite ongoing improvements in resuscitation, care, and outcomes, traumatic injury remains a significant health care and economic burden. The causes are multifactorial, but our approach to the clinical management of these patients remains limited by our current understanding of the pathobiology of the disease. A multicenter, multidisciplinary program known as the "Inflammation and the Host Response to Injury" Large Scale Collaborative Research Program was created by the National Institute of General Medical Sciences (NIGMS, U54 GM062119-10) in 2001 in a 10-year effort to address some of these issues. Its primary goal is to describe the human genomic response to severe trauma and burns, and to examine changes in gene expression in the context of different clinical outcomes. The Program has not only successfully implemented clinical care guidelines for managing the severe trauma patient based on the best available evidence to minimize iatrogenic variability, but it has also examined the genome-wide, immune-inflammatory response in total and isolated blood leukocyte populations. This review will address current milestones as well as future directions for the Program.

4.
J Trauma ; 67(4): 792-801, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19820587

ABSTRACT

INTRODUCTION: As outcome to severe trauma is frequently affected by massive blood loss and consecutive hemorrhagic shock, replacement of red blood cell (RBC) units remains indispensable. Administration of RBC units is an independent risk factor for adverse outcome in patients with trauma. The impact of massive blood transfusion or uncrossmatched blood transfusion on the patients' immune response in the early posttraumatic period remains unclear. MATERIAL: Thirteen patients presenting with blunt multiple injuries (Injury Severity Score >16) were studied. Monocytes were obtained on admission and at 6, 12, 24, 48, and 72 hours after trauma. Biotinylated complementary RNA targets were hybridized to Affymetrix HG U 133A microarrays. The data were analyzed by a supervised analysis based on whether the patients received massive blood transfusions, and then subsequently, by hierarchical clustering, and by Ingenuity pathway analysis. RESULTS: Supervised analysis identified 224 probe sets to be differentially expressed (p < 0.001) in patients who received massive blood transfusion, when compared with those who did not. In addition, 331 probe sets were found differentially expressed (p < 0.001) in patients who received uncrossmatched RBC units in comparison with those who exclusively gained crossmatched ones. Functional pathway analysis of the respectively identified gene expression profiles suggests a contributory role by the AKT/PI3Kinase pathway, the mitogen-activated protein-kinase pathway, the Ubiquitin pathway, and the diverse inflammatory networks. CONCLUSION: We exhibited for the first time a serial, sequential screening analysis of monocyte messenger RNA expression patterns in patients with multiple trauma indicating a strongly significant association between the patients' genomic response in blood monocytes and massive or uncrossmatched RBC substitution.


Subject(s)
Erythrocyte Transfusion , Monocytes/metabolism , Multiple Trauma/genetics , RNA, Messenger/metabolism , Wounds, Nonpenetrating/genetics , Adult , Female , Genome, Human , Humans , I-kappa B Proteins/metabolism , Injury Severity Score , Male , Middle Aged , Multiple Organ Failure/genetics , NF-KappaB Inhibitor alpha , NF-kappa B/antagonists & inhibitors , Oligonucleotide Array Sequence Analysis , Protein Array Analysis , Ubiquitin C/metabolism , Up-Regulation/physiology , Young Adult
5.
J Perinatol ; 29(2): 79-88, 2009 Feb.
Article in English | MEDLINE | ID: mdl-18769381

ABSTRACT

Prevention of neonatal infection-related mortality represents a significant global challenge particularly in the vulnerable premature population. The increased risk of death from sepsis is likely due to the specific immune deficits found in the neonate as compared to the adult. Stimulation of the neonatal immune system to prevent and/or treat infection has been attempted in the past largely without success. In this review, we identify some of the known deficits in the neonatal immune system and their clinical impact, summarize previous attempts at immunomodulation and the outcomes of these interventions, and discuss the potential of novel immunomodulatory therapies to improve neonatal sepsis outcome.


Subject(s)
Immunologic Factors/therapeutic use , Sepsis/drug therapy , Sepsis/prevention & control , Female , Humans , Infant, Newborn , Male , Probiotics/therapeutic use , Risk Factors , Sepsis/immunology
6.
Virology ; 384(1): 151-60, 2009 Feb 05.
Article in English | MEDLINE | ID: mdl-19056101

ABSTRACT

Intratracheal (i.t.) infection of mice with cowpox virus (CPXV), is lethal at a lower dose than intranasal (i.n.) inoculation. CPXV deleted for cytokine response modifier A (CPXVDeltacrmA) was attenuated compared to CPXV after i.t. inoculation. This attenuation could not be attributed to differences in virus replication, immunomodulators, or cells infiltrating the lungs. Deletion of crmA also caused attenuation during intradermal (i.d.) infection. In contrast to i.t.-inoculated virus, deletion of crmA reduced virus replication at the site of infection. This difference correlated to increased numbers of CD3(+) cells in CPXVDeltacrmA-associated dermal lesions. Thus, crmA is a virulence factor in mice during either pulmonary or dermal cowpox infection; however the influence of crmA is more evident during i.d. inoculation. This suggests that the host immune response differs in the two routes of infection and emphasizes the need to consider the effect of route of infection when examining functions of virulence factors in vivo.


Subject(s)
Cowpox virus/genetics , Cowpox/veterinary , Serpins/genetics , Viral Proteins/genetics , Animals , Caspases/metabolism , Cowpox/enzymology , Cowpox/mortality , Cowpox/physiopathology , Cowpox virus/pathogenicity , Gene Expression Regulation, Viral , Humans , Mice , Mice, Inbred C57BL , Smallpox/virology , Species Specificity , Virulence
7.
Scand J Immunol ; 68(6): 565-71, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18959626

ABSTRACT

Recent studies have shown that increased lymphocyte apoptosis contributes to sepsis-induced mortality. Furthermore, studies have demonstrated that IL-10 can suppress lymphocyte apoptosis, in part, by upregulating Bcl-2 expression and interfering with activation induced cell death. We have previously shown that intrathymic delivery of IL-10 with an adenoviral vector in wild-type mice significantly improves outcome to sepsis. Presently, we investigated the role of endogenous IL-10 expression on thymocyte apoptosis and outcome in IL-10 null mice subject to induction of generalized polymicrobial peritonitis via cecal ligation and puncture. Compared to wild-type C57BL/6 mice, IL-10 null mice demonstrated increased mortality and enhanced lymphocyte apoptosis. Intrathymic injection with an adenoviral vector expressing human IL-10 prior to cecal ligation and puncture in IL-10 null mice significantly improved outcome and decreased thymic caspase-3 activity. Furthermore, plasma concentrations of IL-6 were also significantly reduced in IL-10 null mice treated with the IL-10 expressing adenovirus. In contrast, injection of a control adenovirus did not improve outcome in IL-10 null mice, nor was caspase-3 activity reduced. Thus, local thymic expression of IL-10 not only improves outcome but also reduces local tissue apoptosis and caspase-3 activity, and appears to attenuate the systemic proinflammatory cytokine response.


Subject(s)
Apoptosis/immunology , Interleukin-10/immunology , Sepsis/therapy , Thymus Gland/immunology , Adenoviridae/genetics , Animals , Caspase 3/immunology , Caspase 3/metabolism , Disease Models, Animal , Female , Gene Expression , Genetic Therapy , Humans , Interleukin-10/genetics , Interleukin-10/metabolism , Interleukin-6/blood , Lymphocytes/immunology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Sepsis/immunology
8.
Langenbecks Arch Surg ; 392(3): 255-65, 2007 May.
Article in English | MEDLINE | ID: mdl-17404753

ABSTRACT

BACKGROUND: Posttraumatic immune system activation in major trauma patients is linked to systemic inflammatory response syndrome, multiple organ failure (MOF), and mortality. Recent studies suggest that genome-wide expression is altered in response to distinct clinical parameters; however, the functional allocation of theses genes remains unclear. PATIENTS AND METHODS: Thirteen patients after major trauma (Injury Severity Score < 16) were studied. Monocytes were obtained on admission (within 90 min) and at 6, 12, 24, 48, and 72 h after trauma. Complementary ribonucleic acid (RNA) targets were hybridized to Affymetrix HG U 133A microarrays. Searching for genes that are differentially expressed, the patients were dichotomously assigned depending upon survival, injury severity, and MOF. The data were analyzed by supervised analysis, clustering, and comparative pathway analysis. RESULTS: Gene expression profiles of patients with adverse outcomes (763 probe sets) mainly consist of those involved in "immunological activation" or "cellular movement," whereas the gene set associated with MOF (660) is associated with "cancer" and "cell death." Injury severity (295) leads to an overexpression of genes involved in inflammatory disease. CONCLUSION: We demonstrate for the first time a serial, sequential screening analysis of monocyte messenger RNA expression patterns after multiple injury indicating a strongly significant connection between the patients' expression profile and different clinical parameters. The latter provoke a characteristic overexpression of specific functional gene ontologies. Further studies to clarify clinical consequence of this differential gene regulation are currently anticipated.


Subject(s)
Gene Expression Profiling , Multiple Organ Failure/genetics , Multiple Trauma/genetics , Oligonucleotide Array Sequence Analysis , Trauma Severity Indices , Adult , Aged , Female , Genome, Human , Humans , Male , Middle Aged , Monocytes , RNA, Messenger/analysis
9.
Gene Ther ; 13(3): 276-82, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16251998

ABSTRACT

Targeted expression of interleukin-10 (IL-10) has been proposed as a means to suppress acute and chronic inflammation. We explored the capacity of targeted adenoviral expression of human or viral IL-10 to improve outcome in a zymosan-induced model of acute lung injury and multisystem organ failure. Intratracheal administration of adenovirus expressing either human or viral IL-10 prior to zymosan administration significantly improved survival at a dose of 10(7) particles (P<0.01), whereas the same recombinant vectors were ineffective at 10(8) particles and increased mortality at 10(9) particles. Improved survival after administration of 10(7) particles of adenovirus expressing viral or human IL-10 was associated with local tissue expression of IL-10 (100-300 pg/g wet wt). In contrast, mortality after administration of 10(9) particles was associated with markedly elevated IL-10 expression, both in the lung (10000-70000 pg/g wet wt) and systemically (1000-3000 pg/ml plasma), with evidence of an exaggerated systemic inflammatory response (plasma IL-6 and TNFalpha). Targeted gene expression of IL-10 can be used to treat acute inflammatory processes, but increased doses resulting in its systemic release are not associated with improvements in outcome, and may actually exacerbate acute inflammatory processes.


Subject(s)
Adenoviridae/genetics , Genetic Therapy/methods , Genetic Vectors/administration & dosage , Immunotherapy, Active/methods , Interleukin-10/genetics , Multiple Organ Failure/therapy , Acute Disease , Animals , Bacterial Infections/immunology , Dose-Response Relationship, Immunologic , Gene Expression , Gene Targeting , Humans , Interleukin-10/immunology , Interleukin-6/blood , Lung/immunology , Lung Diseases/immunology , Mice , Mice, Inbred C57BL , Models, Animal , Multiple Organ Failure/immunology , Multiple Organ Failure/mortality , Polysaccharides, Bacterial , Recombinant Fusion Proteins/administration & dosage , Recombinant Fusion Proteins/genetics , Survival Rate , Transduction, Genetic/methods , Tumor Necrosis Factor-alpha/analysis , Zymosan
10.
J Autoimmun ; 24(4): 297-310, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15927792

ABSTRACT

Autocrine granulocyte macrophage-colony stimulating factor (GM-CSF) sequentially activates intracellular components in monocyte/macrophage production of the pro-inflammatory and immunoregulatory prostanoid, prostaglandin E2 (PGE2). GM-CSF first induces STAT5 signaling protein phosphorylation, then prostaglandin synthase 2 (COX2/PGS2) gene expression, and finally IL-10 production, to downregulate the cascade. Without activation, monocytes of at-risk, type 1 diabetic (T1D), and autoimmune thyroid disease (AITD) humans, and macrophages of nonobese diabetic (NOD) mice have aberrantly high GM-CSF, PGS2, and PGE2 expression, but normal levels of IL-10. After GM-CSF stimulation, repressor STAT5A and B isoforms (80-77kDa) in autoimmune human and NOD monocytes and activator STAT5A (96-94kDa) and B (94-92kDa) isoforms in NOD macrophages stay persistently tyrosine phosphorylated. This STAT5 phosphorylation persisted despite treatment in vitro with IL-10, anti-GM-CSF antibody, or the JAK2/3 inhibitor, AG490. Phosphorylated STAT5 repressor isoforms in autoimmune monocytes had diminished DNA binding capacity on GAS sequences found in the PGS2 gene enhancer. In contrast, STAT5 activator isoforms in NOD macrophages retained their DNA binding capacity on these sites much longer than in healthy control strain macrophages. These findings suggest that STAT5 dysfunction may contribute to dysregulation of GM-CSF signaling and gene activation, including PGS2, in autoimmune monocytes and macrophages.


Subject(s)
Autoimmune Diseases/immunology , DNA-Binding Proteins/immunology , Macrophages/immunology , Milk Proteins/immunology , Monocytes/immunology , Signal Transduction/immunology , Trans-Activators/immunology , Adolescent , Adult , Animals , Autoimmune Diseases/pathology , Cells, Cultured , Child , Female , Gene Expression Regulation/immunology , Humans , Macrophages/pathology , Male , Mice , Mice, Inbred NOD , Middle Aged , Monocytes/pathology , STAT5 Transcription Factor , Transcriptional Activation , Tumor Suppressor Proteins
11.
Scand J Immunol ; 61(4): 309-15, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15853912

ABSTRACT

Dendritic cells (DC) represent a potential target for gene therapy. In their ability to process antigens and present them to T cells, DC have been allocated a unique role as initiators of the immune response in both the innate and acquired immunity. Recent in vitro studies have showed the feasibility of DC transduction with adenoviral recombinants. In cancer therapy, targeting of DC with adenovirus has been proved to be effective in inhibiting tumour growth, as well as in reducing the number of tumour metastases. The aim of our study is to evaluate the feasibility of in vivo transduction of DC in a murine lymphocyte-rich compartment (thymus) as a potential treatment for acute inflammatory diseases. Nearly 50% of the total thymic DC were transduced with a first-generation adenoviral construct following intrathymic injection, and post-transductional inflammation was neglectable. Transduction of thymic cells with adenoviral recombinants was able to induce the expression of an intracellular protein (beta-galactosidase, green fluorescent protein), as well as the secretion of human interleukin-10, within the local compartment. Furthermore, this induction of the latter significantly decreased thymic apoptosis in the applied model of acute bacterial peritonitis (cecal ligation and puncture).


Subject(s)
Adenoviridae/genetics , Dendritic Cells/immunology , Immunotherapy, Adoptive/methods , Thymus Gland/immunology , Animals , Caspase 3 , Caspases/immunology , Dendritic Cells/physiology , Female , Flow Cytometry , Genetic Vectors/genetics , Green Fluorescent Proteins/biosynthesis , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/immunology , Interleukin-10/biosynthesis , Interleukin-10/blood , Interleukin-10/genetics , Interleukin-10/metabolism , Interleukin-6/blood , Mice , Mice, Inbred C57BL , Sepsis/immunology , Specific Pathogen-Free Organisms , Thymus Gland/cytology , Thymus Gland/physiology , Transduction, Genetic , Transgenes , Tumor Necrosis Factor-alpha/immunology , beta-Galactosidase/biosynthesis , beta-Galactosidase/genetics , beta-Galactosidase/immunology
12.
Eur Surg Res ; 36(5): 266-73, 2004.
Article in English | MEDLINE | ID: mdl-15359089

ABSTRACT

BACKGROUND: Since the plasma cytokine profile reflects the body's inflammatory response to injury, this study was designed to prospectively observe the plasma cytokine levels in response to the degree of different sorts of abdominal surgical trauma. METHODS: Plasma levels of TNF-alpha, type I TNF receptor (p55), type II TNF receptor (p75), IL-6, IL-8, IL-10, phospholipase A(2) (PLA(2)), and haptoglobin were measured peri-operatively in patients undergoing bowel resection for inflammatory bowel disease or diverticulitis (IBD) (n = 9), elective repair of abdominal aortic aneurysm (AAA) (n = 9), or laparoscopic cholecystectomy (lap chole) (n = 9). RESULTS: The IBD patients showed a significant (p < 0.05) post-operative elevation in plasma IL-6, p55, p75, and PLA(2) levels, but no significant change in TNF-alpha, IL-8, IL-10 or haptoglobin levels. The AAA patients had a significant post-operative rise in IL-10 levels and a significant decrease in plasma haptoglobin levels, but no significant change of TNF-alpha, IL-6, IL-8, p55, p75, or PLA(2) concentrations. The lap chole patients demonstrated no significant change in any of these parameters. CONCLUSION: These data show that IL-6, IL-10, p55, and p75 are markers to measure the degree of inflammatory stress associated with abdominal operative procedures and demonstrate the relative lack of a cytokine response to laparoscopic cholecystectomy.


Subject(s)
Aortic Aneurysm, Abdominal/surgery , Digestive System Surgical Procedures/adverse effects , Inflammation/etiology , Inflammatory Bowel Diseases/surgery , Vascular Surgical Procedures/adverse effects , Adult , Aged , Aortic Aneurysm, Abdominal/blood , Female , Haptoglobins/metabolism , Humans , Inflammatory Bowel Diseases/blood , Interleukins/blood , Male , Middle Aged , Phospholipases A/blood , Postoperative Period , Prospective Studies , Receptors, Tumor Necrosis Factor, Type I/blood , Receptors, Tumor Necrosis Factor, Type II/blood
13.
J Autoimmun ; 22(3): 227-33, 2004 May.
Article in English | MEDLINE | ID: mdl-15041043

ABSTRACT

Aberrant prostaglandin synthase 2 (PGS2/COX2) expression constitutes an antigen presenting cell (APC) dysfunction seen in monocytes of humans at risk for or with Type 1 diabetes. During endotoxin activation of PGS2 expression in healthy monocytes, granulocyte-monocyte colony stimulating factor (GM-CSF) is activated and, in turn, promotes PGS2 gene activation. GM-CSF is considered a major target the action for IL10 in its suppression of PGS2. We found that the PGS2 expression in monocytes from 47% of at-risk and diabetic humans tested were highly resistant to suppression by IL10 (maintaining > or =50% of their untreated expression), and had significantly increased GM-CSF production in vitro (1043+/-SD2798 pg/10(6)cells, subject n=35, vs 29.7+/-SD91 pg/10(6)cells, control n=20; P=0.0165). The PGS2 insensitivity to IL10 of these cells was not due to a lack of IL10 functionality or its suppression of GM-CSF. In contrast to its effects on PGS2, IL10 regulation of GM-CSF and other monocyte factors (i.e., DR, IL1beta, TNFalpha, IL12, CD54, and CD64) remained intact. These findings suggest that the inability of IL10 to properly downregulate PGS2 gene expression may contribute to its dysregulation in Type 1 diabetes.


Subject(s)
Diabetes Mellitus, Type 1/metabolism , Interleukin-10/metabolism , Isoenzymes/genetics , Monocytes/metabolism , Prostaglandin-Endoperoxide Synthases/genetics , Adolescent , Adult , Aged , Child , Child, Preschool , Cyclooxygenase 2 , Diabetes Mellitus, Type 1/enzymology , Diabetes Mellitus, Type 1/immunology , Granulocyte-Macrophage Colony-Stimulating Factor/biosynthesis , Humans , Isoenzymes/biosynthesis , Membrane Proteins , Middle Aged , Monocytes/immunology , Prostaglandin-Endoperoxide Synthases/biosynthesis
14.
J Endotoxin Res ; 7(5): 375-80, 2001.
Article in English | MEDLINE | ID: mdl-11753206

ABSTRACT

Lipopolysaccharide and D-galactosamine induced lethality and apoptotic liver injury is dependent upon endogenously produced TNF-alpha. Unlike the response to high dose lipopolysaccharide alone, death in this model is a direct result of hepatocyte apoptosis. In a series of recent studies, we have demonstrated that mortality and hepatic injury following lipopolysaccharide administration in D-galactosamine-sensitized mice is dependent upon secreted 17 kDa TNF-alpha acting primarily through the p55 TNF receptor. Transgenic mice expressing null forms of TNF-alpha, the p55 receptor, or expressing only a cell-associated form of TNF-alpha exhibited no mortality and only modest liver injury when challenged with 8 mg of D-galactosamine and 100 ng of lipopolysaccharide. Although Fas ligand expression is increased in the liver, it appeared to play no significant role in outcome, since mice expressing a mutant form of Fas ligand are still sensitive to LPS- and D-galactosamine-induced lethality. Finally, we have seen significant variation in LPS- and D-galactosamine-mediated lethality among different strains of mice. The non-obese diabetic or NOD mouse is highly resistant to LPS-and D-galactosamine-induced lethality, and this appears to be secondary to a post-receptor defect in p55 TNF receptor signaling. The studies confirm an essential role for TNF-alpha and p55 TNF receptor signaling in the hepatocyte apoptosis and lethality associated with lipopolysaccharide and D-galactosamine administration.


Subject(s)
Apoptosis/genetics , Chemical and Drug Induced Liver Injury , Galactosamine/pharmacology , Lipopolysaccharides/pharmacology , Longevity/genetics , Tumor Necrosis Factor-alpha/genetics , Animals , Caspase 3 , Caspases/biosynthesis , Drug Therapy, Combination , Humans , Liver/drug effects , Liver/enzymology , Liver/pathology , Liver Diseases/blood , Liver Diseases/pathology , Longevity/drug effects , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, Knockout , Mice, Transgenic , Necrosis , Recombinant Proteins , Species Specificity , Tumor Necrosis Factor-alpha/biosynthesis , Tumor Necrosis Factor-alpha/pharmacology
15.
J Appl Physiol (1985) ; 91(5): 2213-23, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11641364

ABSTRACT

Pharmacokinetics and immunogenicity of six different recombinant human soluble p55 tumor necrosis factor (TNF) receptor I (sTNFR-I) constructs were evaluated in juvenile baboons. The constructs included either an sTNFR-I IgG1 immunoadhesin (p55 sTNFR-I Fc) or five different sTNFR-I constructs covalently linked to polyethylene glycol. The constructs were administered intravenously three times, and pharmacokinetics and immunogenicity were examined over 63 days. All of the constructs were immunogenic, with the exception of a 2.6-domain monomeric sTNFR-I. To evaluate whether the nonimmunogenic 2.6-domain monomeric construct could protect baboons against TNF-alpha-induced mortality, baboons were pretreated with 1, 5, or 10 mg/kg body wt and were compared with baboons receiving either placebo or 1 mg/kg body wt of the dimeric 4.0-domain sTNFR-I construct (n = 3 each) before lethal Escherichia coli bacteremia. The monomeric construct protected baboons and neutralized TNF bioactivity, although greater quantities were required compared with the dimeric 4.0-domain sTNFR-I construct. We conclude that E. coli-recombinant-derived human sTNFR-I constructs can be generated with minimal immunogenicity on repeated administration and still protect against the consequences of exaggerated TNF-alpha production.


Subject(s)
Immunoglobulin G/metabolism , Papio/metabolism , Receptors, Tumor Necrosis Factor/metabolism , Amino Acid Sequence , Animals , Blood Cell Count , Cloning, Molecular , Cytokines/metabolism , Enzyme-Linked Immunosorbent Assay , Escherichia coli/genetics , Etanercept , Half-Life , Hemodynamics/physiology , Humans , Immunoglobulin G/immunology , Kinetics , Molecular Sequence Data , Polyethylene Glycols , Receptors, Tumor Necrosis Factor/immunology , Tumor Necrosis Factor-alpha/pharmacokinetics , Tumor Necrosis Factor-alpha/pharmacology
16.
Proc Natl Acad Sci U S A ; 98(20): 11503-8, 2001 Sep 25.
Article in English | MEDLINE | ID: mdl-11553765

ABSTRACT

Sepsis remains a significant clinical conundrum, and recent clinical trials with anticytokine therapies have produced disappointing results. Animal studies have suggested that increased lymphocyte apoptosis may contribute to sepsis-induced mortality. We report here that inhibition of thymocyte apoptosis by targeted adenovirus-induced thymic expression of human IL-10 reduced blood bacteremia and prevented mortality in sepsis. In contrast, systemic administration of an adenovirus expressing IL-10 was without any protective effect. Improvements in survival were associated with increases in Bcl-2 expression and reductions in caspase-3 activity and thymocyte apoptosis. These studies demonstrate that thymic apoptosis plays a critical role in the pathogenesis of sepsis and identifies a gene therapy approach for its therapeutic intervention.


Subject(s)
Apoptosis/drug effects , Interleukin-10/genetics , Sepsis/therapy , Animals , Disease Models, Animal , Female , Humans , Interleukin-10/therapeutic use , Mice , Mice, Inbred C57BL , Sepsis/immunology
17.
Surgery ; 130(2): 363-9, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11490372

ABSTRACT

BACKGROUND: Chemoresistance may involve the anti-apoptotic transcriptional regulator, nuclear factor-kappa B (NF-kappa B). The purpose of this study was to determine whether chemotherapy induces NF-kappa B activation in a human colon cancer cell line (SW48) and whether NF-kappa B is constitutively activated in colorectal cancer. METHODS: SW48 cells were incubated with gemcitabine hydrochloride (Gemzar) in the presence and absence of the 26s proteasome inhibitor, MG132, and NF-kappa B binding (electrophoretic mobility shift assay), DNA synthesis (tritiated thymidine uptake), cell viability (3-[4,5-dimethylthiazol-2-yl]-diphenyl-tetrazolium bromide assay), and apoptosis (caspase-3 activity) were measured at 24 hours. NF-kappa B binding (electrophoretic mobility shift assay) was also assayed in 10 colorectal cancer tumors. RESULTS: SW48 cells demonstrated constitutive NF-kappa B binding that was enhanced by gemcitabine hydrochloride in a dose-dependent manner. MG132 inhibited NF-kappa B binding and enhanced gemcitabine hydrochloride's inhibition of DNA synthesis (gemcitabine hydrochloride = 73% +/- 1.4% vs gemcitabine hydrochloride + MG132 = 6% +/- 0.4%, P <.05), cell killing (gemcitabine hydrochloride = 87% +/- 2.0 vs gemcitabine hydrochloride + MG132 = 25% +/- 1.3%, P <.05), and caspase-3 activity (gemcitabine hydrochloride = 870 +/- 17.4 vs gemcitabine hydrochloride + MG132 = 1075 +/- 20.4, P <.05). NF-kappa B binding was increased in 8 of 10 colorectal cancer tumors compared with adjacent normal mucosa. CONCLUSIONS: Gemcitabine hydrochloride enhances NF-kappa B binding in a colorectal cancer cell line, whereas inhibition of NF-kappa B enhances gemcitabine hydrochloride's antitumor activity. NF-kappa B is also activated in human colorectal cancer. NF-kappa B may identify chemoresistant tumors, whereas inhibition of NF-kappa B may be a novel, biologically based therapy. (Surgery 2001;130:363-9).


Subject(s)
Colorectal Neoplasms/pathology , NF-kappa B/metabolism , Antimetabolites, Antineoplastic/toxicity , Caspase 3 , Caspases/metabolism , Colon/metabolism , Colon/pathology , Colorectal Neoplasms/metabolism , Cysteine Proteinase Inhibitors/pharmacology , DNA/biosynthesis , Deoxycytidine/analogs & derivatives , Deoxycytidine/toxicity , Humans , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Leupeptins/pharmacology , NF-kappa B/antagonists & inhibitors , Protein Binding/drug effects , Protein Binding/physiology , Rectum/metabolism , Rectum/pathology , Tumor Cells, Cultured , Gemcitabine
18.
Shock ; 16(2): 83-96, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11508871

ABSTRACT

An intact innate and acquired immune response are essential for defeating systemic microbial infections. Recognition molecules, inflammatory cells, and the cytokines they produce are the principal means for host tissues to recognize invading microbes and to initiate intercellular communication between the innate and acquired immune systems. However, activation of host innate immunity may also occur in the absence of microbial recognition, through expression of internal "danger" signals produced by tissue ischemia and necrosis. When activation of the innate immune system is severe enough, the host response itself can propel the patient into a systemic inflammatory response syndrome (SIRS), or even multiple system organ failure (MSOF) and shock. Although most patients survive the initial SIRS insult, these patients remain at increased risk of developing secondary or opportunistic infections because of the frequent onset of a compensatory anti-inflammatory response syndrome (CARS). The initial activation of the innate immune response often leads to macrophage deactivation, T-cell anergy, and the rapid apoptotic loss of lymphoid tissues, which all contribute to the development of this CARS syndrome and its associated morbidity and mortality. Initial efforts to treat the septic patient with anticytokine therapies directed at the SIRS response have been disappointing, and therapeutic efforts to modify the immune response during sepsis syndromes will require a more thorough understanding of the innate and acquired immune responses and the increased apoptosis in the lymphoid tissue.


Subject(s)
Immunity, Innate , Systemic Inflammatory Response Syndrome/immunology , Humans , Immunity , Models, Immunological
19.
J Immunol ; 167(2): 1053-9, 2001 Jul 15.
Article in English | MEDLINE | ID: mdl-11441115

ABSTRACT

Adenovirus (Ad) gene therapy has been proposed as a drug-delivery system for the targeted administration of protein-based therapies, including growth factors and biological response modifiers. However, inflammation associated with Ad transduction has raised concern about its safety and efficacy in acute inflammatory diseases. In the present report, intratracheal and i.v. administration of a first-generation adenoviral recombinant (E1,E3 deleted) either containing an empty cassette or expressing the anti-inflammatory cytokines viral or human IL-10 (IL-10) was administered to mice subjected to zymosan-induced multisystem organ failure or to acute necrotizing pancreatitis. Pretreatment of mice with the intratracheal instillation of Ad expressing human IL-10 or viral IL-10 reduced weight loss, attenuated the proinflammatory cytokine response, and reduced mortality in the zymosan-induced model, whereas pretreatment with a control adenoviral recombinant did not significantly exacerbate the response. Pretreatment of mice with pancreatitis using adenoviral vectors expressing IL-10 significantly reduced the degree of pancreatic and liver injury and liver inflammation when administered systemically, but not intratracheally. We conclude that adenoviral vectors can be administered prophylactically in acute inflammatory syndromes, and expression of the anti-inflammatory protein IL-10 can be used to suppress the underlying inflammatory process.


Subject(s)
Adenoviridae/genetics , Genetic Therapy/methods , Interleukin-10/administration & dosage , Interleukin-10/genetics , Sepsis/immunology , Sepsis/therapy , Viral Proteins/administration & dosage , Viral Proteins/genetics , Animals , Antimetabolites/toxicity , Choline Deficiency/genetics , Choline Deficiency/immunology , Choline Deficiency/pathology , Choline Deficiency/therapy , Cytokines/blood , Cytokines/metabolism , Ethionine/toxicity , Female , Genetic Vectors/administration & dosage , Genetic Vectors/immunology , Humans , Injections, Intravenous , Intubation, Intratracheal , Liver/immunology , Liver/pathology , Lung/immunology , Lung/pathology , Mice , Mice, Inbred C57BL , Pancreatitis/genetics , Pancreatitis/immunology , Pancreatitis/pathology , Pancreatitis/therapy , Sepsis/genetics , Sepsis/pathology , Zymosan/toxicity
20.
Regul Pept ; 99(2-3): 69-77, 2001 Jun 15.
Article in English | MEDLINE | ID: mdl-11384767

ABSTRACT

The weight-reducing effects of leptin are predominantly mediated through the hypothalamus in the brain. Gene therapy strategies designed for weight control have so far tested the short-term effect of peripherally delivered viral vectors encoding the leptin gene. In order to circumvent the multiple peripheral effects of hyperleptinemia and to overcome the age-related development of leptin resistance due to multiple factors, including defective leptin transport across the blood brain barrier, we determined whether delivery of viral vectors directly into the brain is a viable therapeutic strategy for long-term weight control in normal wild-type rats. A recombinant adeno-associated virus (rAAV) vector encoding rat leptin (Ob) cDNA was generated (rAAV-betaOb). When administered once intracerebroventricularly (i.c.v.), rAAV-betaOb suppressed the normal time-related weight gain for extended periods of time in adult Sprague-Dawley rats. The vector expression was confirmed by immunocytochemical localization of GFP and RT-PCR analysis of leptin in the hypothalamus. This sustained restraint on weight gain was not due to shifts in caloric consumption because food-intake was similar in rAAV-betaOb-treated and rAAV-GFP-treated control rats throughout the experiment. Weight gain suppression, first apparent after 2 weeks, was a result of reduced white fat depots and was accompanied by drastically reduced serum leptin and insulin concentrations in conjunction with normoglycemia. Additionally, there was a marked increase in uncoupling protein-1 (UCP1) mRNA expression in brown adipose tissue, thereby indicating increased energy expenditure through thermogenesis. Seemingly, a selective enhancement in energy expenditure following central delivery of the leptin gene is a viable therapeutic strategy to control the age-related weight gain and provide protection from the accompanying multiple peripheral effects of hyperleptinemia and hyperinsulinemia.


Subject(s)
Adipose Tissue/metabolism , Feeding Behavior , Insulin Antagonists/administration & dosage , Insulin/blood , Leptin/genetics , Obesity/prevention & control , Weight Gain/genetics , Adenoviridae/genetics , Adipose Tissue/physiopathology , Adipose Tissue, Brown/metabolism , Aging/blood , Aging/genetics , Aging/physiology , Animals , Body Composition/genetics , Carrier Proteins/metabolism , Female , Genetic Therapy/methods , Genetic Vectors/administration & dosage , Genetic Vectors/genetics , Green Fluorescent Proteins , Hypothalamus/metabolism , Immunohistochemistry , Injections, Intraventricular , Ion Channels , Leptin/administration & dosage , Leptin/biosynthesis , Leptin/blood , Luminescent Proteins/analysis , Membrane Proteins/metabolism , Mitochondrial Proteins , Obesity/genetics , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Uncoupling Protein 1
SELECTION OF CITATIONS
SEARCH DETAIL
...