Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 104
Filter
1.
Eur Respir J ; 24(1): 107-15, 2004 Jul.
Article in English | MEDLINE | ID: mdl-15293612

ABSTRACT

Lung expression of the extracellular matrix protein, laminin, and its receptor, laminin-1 receptor, were examined in a mouse model of asthma with airway remodelling. Ovalbumin (OVA) was administered to BALB/c mice, intraperitoneally on days 0 and 14, and intranasally periodically between days 14 and 75. The mice developed airway eosinophil and mononuclear inflammatory cell infiltration and fibrosis. On day 76, a marked increase in total laminin was seen in the airways of OVA-treated mice compared to controls by Western blot analysis. The increased laminin expression was detected immunocytochemically in the thickened subepithelial basement membrane and around airways and blood vessels. The OVA-treated mice showed increased expression of the alpha1, beta1, and gamma1 chains of the laminin-1 isoform in monocytes, macrophages and eosinophils infiltrating the airways. Laminin-1 receptor expression was increased in inflammatory and endothelial cells in the lungs of OVA-treated mice compared to controls. Treatment of OVA-sensitised/challenged mice with dexamethasone reduced airway expression of laminin and laminin-1 receptor in OVA-treated mice but not airway hyperresponsiveness to methacholine. Laminin deposition may be an important component of the airway remodelling observed in chronic allergic lung inflammation and is a process modulated by corticosteroids.


Subject(s)
Asthma/immunology , Bronchial Hyperreactivity/immunology , Dexamethasone/pharmacology , Eosinophilia/drug therapy , Receptors, Laminin/immunology , Receptors, Laminin/metabolism , Airway Resistance/drug effects , Animals , Asthma/metabolism , Biomarkers/analysis , Biopsy, Needle , Blotting, Western , Bronchial Hyperreactivity/metabolism , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Administration Schedule , Eosinophilia/diagnosis , Female , Immunohistochemistry , Mice , Mice, Inbred BALB C , Risk Factors , Sensitivity and Specificity
2.
J Ultrasound Med ; 20(5): 473-80, 2001 May.
Article in English | MEDLINE | ID: mdl-11345104

ABSTRACT

OBJECTIVE: To develop a noninvasive method for treatment of uterine fibroid tumors using high-intensity focused ultrasound. Optimal high-intensity focused ultrasound treatment would be dependent on quantitative information about ultrasonic tissue characteristics. METHODS: Ultrasonic attenuation and the sound speed of fresh human fibroid tumors and myometrium were measured as a function of frequency (1-3 MHz) by using a pulse transmission technique before and after in vitro high-intensity focused ultrasound treatment (3.5 MHz at an intensity of 2,000 W/cm2). RESULTS: The ranges of the attenuation coefficients, before and after high-intensity focused ultrasound treatment, were 0.9 to 2.2 and 1.8 to 3.9 dB/cm2, respectively, for fibroid tumors and 0.5 to 1.6 and 1.7 to 3.3 dB/cm2, respectively, for myometrium. Although the sound speed appeared to be independent of frequency (1,611 to 1,616 m/s at 1 to 3 MHz) in both types of tissues, a slight increase of approximately 4 to 14 m/s was observed after high-intensity focused ultrasound treatment. CONCLUSIONS: The results of this study represent our first reported values of the attenuation coefficient and sound speed in fibroid tumors and myometrium before and after high-intensity focused ultrasound treatment.


Subject(s)
Leiomyoma/diagnostic imaging , Myometrium/diagnostic imaging , Uterine Neoplasms/diagnostic imaging , Acoustics , Female , Humans , Leiomyoma/therapy , Ultrasonic Therapy , Ultrasonography , Uterine Neoplasms/therapy
3.
Blood ; 97(1): 147-53, 2001 Jan 01.
Article in English | MEDLINE | ID: mdl-11133754

ABSTRACT

Cyclic neutropenia (CN) is a congenital hematopoietic disorder characterized by remarkably regular oscillations of blood neutrophils from near normal to extremely low levels at 21-day intervals. Recurring episodes of severe neutropenia lead to repetitive and sometimes life-threatening infections. To investigate the cellular mechanism of CN, the ultrastructure and the proliferative and survival characteristics of bone marrow-derived CD34(+) early progenitors, CD33(+)/CD34(-) myeloid progenitors, and CD15(+) neutrophil precursors from CN patients and healthy volunteers were studied. The ultrastructural studies showed profound apoptotic features in bone marrow progenitor cells in CN. Colony-forming assays demonstrated a 75% decrease in the number of early myeloid-committed colonies compared with controls. Long-term culture-initiating cell assays demonstrated a 6-fold increase in production of primitive progenitor cells in CN. To determine whether accelerated apoptosis might account for the underproduction of myeloid progenitors, the hematopoietic subpopulations were labeled with fluorescein isothiocyanate-annexin V and analyzed by flow cytometry. Short-term culture of CN cells resulted in apoptosis of approximately 65% of CD34(+) cells, 80% of CD33(+)/CD34(-) cells, and more than 70% of CD15(+) cells, as compared with 20%, 7%, and 15% apoptosis in respective control subpopulations. Evidence of accelerated apoptosis of bone marrow progenitor cells was observed in all 8 patients participating in the study, regardless of the stage in the CN cycle in which bone marrow aspirations were obtained. Granulocyte colony-stimulating factor therapy of CN patients significantly improved survival of bone marrow progenitor cells. These data indicate that ineffective production of neutrophils is due to accelerated apoptosis of bone marrow myeloid progenitor cells in CN.


Subject(s)
Bone Marrow Cells/pathology , Hematopoietic Stem Cells/pathology , Neutropenia/etiology , Annexin A5/metabolism , Antigens, CD34/physiology , Apoptosis/drug effects , Cell Culture Techniques , Cell Division/drug effects , Cell Survival/physiology , Colony-Forming Units Assay , Family Health , Granulocyte Colony-Stimulating Factor/pharmacology , Hematopoiesis , Humans , Microscopy, Electron , Myeloid Progenitor Cells/pathology , Neutropenia/congenital , Neutropenia/pathology , Neutrophils/pathology , Periodicity
4.
Pediatr Res ; 49(1): 30-7, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11134488

ABSTRACT

Escherichia coli K1 is an important neonatal pathogen that is usually transferred from maternal to infant gastrointestinal tract at the time of parturition. Approximately 20% of neonates are colonized, and a proportion of colonized infants goes on to have systemic infection. Entry into the bloodstream from the gastrointestinal tract is hypothesized to occur via epithelial cell invasion. Invasion of multiple epithelial cell lines was studied using gentamicin protection assays and transcytosis of polarized monolayers. Electron microscopy was used to confirm cellular invasion. Cell lines used include two human gastrointestinal lines, Caco-2 and T84; a human respiratory cell line, A549; a human laryngeal cell line, HEp-2; and a canine kidney cell line, MDCK. A virulent E. coli K1 strain, RS218, readily invaded HEp-2, A549, and T84 cell lines in gentamicin protection assays, but was less invasive into MDCK and Caco-2 cells. RS218 also demonstrated transcytosis of both T84 and Caco-2 cells. Four clinical isolates of E. coli K1 demonstrated levels of transcytosis of T84 cells similar to RS218. Caco-2 invasiveness correlated with length of time in tissue culture with maximum invasiveness demonstrated at 11 d in culture, when cells were polarized and differentiated.


Subject(s)
Escherichia coli/physiology , Intestinal Mucosa/microbiology , Animals , Caco-2 Cells , Cell Line , Colchicine/pharmacology , Cytochalasin D/pharmacology , Humans , Intestinal Mucosa/ultrastructure , Microscopy, Electron , Microtubules/drug effects
5.
Am J Pathol ; 158(1): 153-61, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11141488

ABSTRACT

Activation of the Fas/FasL system induces apoptosis of susceptible cells, but may also lead to nuclear factor kappaB activation. Our goal was to determine whether local Fas activation produces acute lung injury by inducing alveolar epithelial cell apoptosis and by generating local inflammatory responses. Normal mice (C57BL/6) and mice deficient in Fas (lpr) were treated by intranasal instillation of the Fas-activating monoclonal antibody (mAb) Jo2 or an irrelevant control mAb, and studied 6 or 24 hours later using bronchoalveolar lavage (BAL), histopathology, DNA nick-end-labeling assays, and electron microscopy. Normal mice treated with mAb Jo2 had significant increases in BAL protein at 6 hours, and BAL neutrophils at 24 hours, as compared to lpr mice and to mice treated with the irrelevant mAb. Neutrophil recruitment was preceded by increased mRNA expression for tumor necrosis factor-alpha, macrophage inflammatory protein-1alpha, macrophage inflammatory protein-2, macrophage chemotactic protein-1, and interleukin-6, but not interferon-gamma, transforming growth factor-ss, RANTES, eotaxin, or IP-10. Lung sections from Jo2-treated normal mice showed neutrophilic infiltrates, alveolar septal thickening, hemorrhage, and terminal dUTP nick-end-labeling-positive cells in the alveolar septae and airspaces. Type II pneumocyte apoptosis was confirmed by electron microscopy. Fas activation in vivo results in acute alveolar epithelial injury and lung inflammation, and may be important in the pathogenesis of acute lung injury.


Subject(s)
Apoptosis , Epithelial Cells/metabolism , Pulmonary Alveoli/metabolism , fas Receptor/metabolism , Acute Disease , Animals , Antibodies, Monoclonal/pharmacology , Bronchoalveolar Lavage Fluid/chemistry , Bronchoalveolar Lavage Fluid/cytology , Cytokines/genetics , Cytokines/metabolism , Epithelial Cells/cytology , Epithelial Cells/drug effects , Fas Ligand Protein , Gene Expression/drug effects , Leukocyte Count , Lung/drug effects , Lung/pathology , Lung/ultrastructure , Membrane Glycoproteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Microscopy, Electron , Neutrophils/cytology , Pneumonia/metabolism , Pneumonia/pathology , Pneumonia/prevention & control , Pulmonary Alveoli/cytology , Pulmonary Alveoli/drug effects , RNA, Messenger/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , fas Receptor/genetics
6.
J Immunol ; 165(8): 4640-8, 2000 Oct 15.
Article in English | MEDLINE | ID: mdl-11035107

ABSTRACT

Leukotrienes are important mediators of the eosinophilic influx and mucus hypersecretion in the lungs in a murine model of asthma. We used in situ PCR in this model of human asthma to detect lung mRNA for 5-lipoxygenase (5-LO) and 5-LO-activating protein (FLAP), key proteins necessary for leukotriene synthesis. Lung tissue was obtained on day 28 from mice treated with i.p. (days 0 and 14) and intranasal (days 14, 25, 26, and 27) OVA or saline. After fixation, the tissue sections underwent protease- and RNase-free DNase digestion, before in situ RT-PCR using target-specific cDNA amplification. 5-LO and FLAP-specific mRNA was visualized by a digoxigenin detection system, and positive cells were analyzed by morphometry. 5-LO and FLAP-specific mRNA and protein were associated primarily with eosinophils and alveolar macrophages in the airways and pulmonary blood vessels in OVA-sensitized/challenged mice. 5-LO and FLAP protein expression increased on a per-cell basis in alveolar macrophages of OVA-treated mice compared with saline controls. Pulmonary blood vessel endothelial cells were also positive for 5-LO, FLAP mRNA, and protein. 5-LO inhibition significantly decreased 5-LO and FLAP-specific mRNA and protein expression in the lung inflammatory cells and endothelial cells. These studies demonstrate a marked increase in key 5-LO pathway proteins in the allergic lung inflammatory response and an important immunomodulatory effect of leukotriene blockade to decrease 5-LO and FLAP gene expression.


Subject(s)
Arachidonate 5-Lipoxygenase/genetics , Carrier Proteins/genetics , Leukotriene Antagonists , Lipoxygenase Inhibitors , Lung/enzymology , Lung/pathology , Membrane Proteins/genetics , Reverse Transcriptase Polymerase Chain Reaction , 5-Lipoxygenase-Activating Proteins , Animals , Arachidonate 5-Lipoxygenase/biosynthesis , Asthma/enzymology , Asthma/metabolism , Asthma/pathology , Carrier Proteins/biosynthesis , Disease Models, Animal , Endothelium, Vascular/metabolism , Endothelium, Vascular/pathology , Female , Humans , Injections, Intraperitoneal , Lung/metabolism , Macrophages, Alveolar/enzymology , Macrophages, Alveolar/metabolism , Membrane Proteins/biosynthesis , Mice , Mice, Inbred BALB C , Organ Specificity/genetics , Ovalbumin/administration & dosage , Ovalbumin/immunology , RNA, Messenger/biosynthesis , RNA, Messenger/metabolism
7.
Am J Obstet Gynecol ; 183(1): 6-11, 2000 Jul.
Article in English | MEDLINE | ID: mdl-10920300

ABSTRACT

OBJECTIVE: The purpose of this study was to investigate the potential efficacy of high-intensity focused ultrasound for the treatment of uterine fibroid tumors in a nude mouse model. STUDY DESIGN: A total of 60 female athymic nude mice were inoculated subcutaneously with 3 to 5 x 10(6) ELT-3 cells, a uterine fibroid tumor cell line. Tumor size was monitored with transcutaneous caliper measurements. The high-intensity focused ultrasound probe was a concave, single-element, high-power transducer that operated at a frequency of 3.5 MHz. The tumors were treated for 30 to 60 seconds using a high-intensity focused ultrasonic intensity of 2000 W/cm(2), depending on the tumor size. RESULTS: A single high-intensity focused ultrasonic treatment resulted in an average reduction in tumor volume of 91% within 1 month of the treatment. Histologic analysis of tumors treated with high-intensity focused ultrasound showed coagulation necrosis and nuclear fragmentation of tumor cells. CONCLUSION: High-intensity focused ultrasound effectively reduced uterine fibroid tumor size in a nude mouse model. Further studies are needed to assess the in situ response of uterine fibroids to high-intensity focused ultrasonic treatment.


Subject(s)
Disease Models, Animal , Leiomyoma/therapy , Ultrasonic Therapy , Uterine Neoplasms/therapy , Animals , Female , Leiomyoma/pathology , Mice , Mice, Nude , Necrosis , Time Factors , Uterine Neoplasms/pathology
8.
J Immunol ; 164(6): 3360-7, 2000 Mar 15.
Article in English | MEDLINE | ID: mdl-10706731

ABSTRACT

Numerous in vitro and in vivo studies in both animal models and human asthmatics have implicated platelet-activating factor (PAF) as an important inflammatory mediator in asthma. In a murine asthma model, we examined the anti-inflammatory activities of recombinant human PAF-acetylhydrolase (rPAF-AH), which converts PAF to biologically inactive lyso-PAF. In this model, mice sensitized to OVA by i.p. and intranasal (i.n.) routes are challenged with the allergen by i.n. administration. The OVA challenge elicits an eosinophil infiltration into the lungs with widespread mucus occlusion of the airways and results in bronchial hyperreactivity. The administration of rPAF-AH had a marked effect on late-phase pulmonary inflammation, which included a significant reduction in airway eosinophil infiltration, mucus hypersecretion, and airway hyperreactivity in response to methacholine challenge. These studies demonstrate that elevating plasma levels of PAF-AH through the administration of rPAF-AH is effective in blocking the late-phase pulmonary inflammation that occurs in this murine allergen-challenge asthma model. These results suggest that rPAF-AH may have therapeutic effects in patients with allergic airway inflammation.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Asthma/prevention & control , Bronchial Hyperreactivity/prevention & control , Lung/pathology , Phospholipases A/pharmacology , Recombinant Proteins/pharmacology , 1-Alkyl-2-acetylglycerophosphocholine Esterase , Allergens/administration & dosage , Allergens/immunology , Animals , Asthma/enzymology , Asthma/immunology , Asthma/pathology , Bronchial Hyperreactivity/enzymology , Bronchial Hyperreactivity/immunology , Bronchial Hyperreactivity/pathology , Bronchoalveolar Lavage Fluid/chemistry , Cell Movement/drug effects , Cell Movement/immunology , Disease Models, Animal , Eosinophils/drug effects , Eosinophils/immunology , Female , Humans , Interleukin-5/metabolism , Lung/drug effects , Lung/immunology , Macrophages/drug effects , Macrophages/immunology , Methacholine Chloride/administration & dosage , Mice , Mice, Inbred BALB C , Mucus/drug effects , Mucus/metabolism , Phospholipases A/blood , Phospholipases A/immunology , Platelet Activating Factor/immunology , Recombinant Proteins/blood , Recombinant Proteins/immunology
9.
J Immunol ; 164(2): 1086-95, 2000 Jan 15.
Article in English | MEDLINE | ID: mdl-10623860

ABSTRACT

In vitro and in vivo studies, in both animal models and human asthmatics, have implicated IL-4 as an important inflammatory mediator in asthma. In a murine asthma model, we examined the anti-inflammatory activities of soluble IL-4R (sIL-4R). In this model, mice sensitized to OVA by i.p. and intranasal (i.n.) routes are challenged with the allergen by i.n. administration. The OVA challenge elicits an eosinophil infiltration into the lungs, with widespread mucus occlusion of the airways, and results in bronchial hyperreactivity. sIL-4R (0.1-100 microgram) was administered by either i.n. or i.p. routes before OVA challenge in OVA-sensitized mice. Both blood and bronchoalveolar lavage fluid levels of sIL-4R were significantly elevated compared with controls by i.n. delivery of 100 microgram sIL-4R; i.p. delivery of 100 microgram sIL-4R only raised blood levels of sIL-4R. The i.n. administration of 100 microgram sIL-4R before allergen challenge significantly reduced late phase pulmonary inflammation, blocking airway eosinophil infiltration, VCAM-1 expression, and mucus hypersecretion. In contrast, i.p. delivery of 100 microgram sIL-4R inhibited only the influx of eosinophils into the lungs, but not airway mucus release. Furthermore, sIL-4R treatment by either i.n. or i.p. routes did not reduce airway hyperreactivity in response to methacholine challenge. Thus, elevating airway levels of sIL-4R through the administration of exogenous sIL-4R is effective in blocking the late phase pulmonary inflammation that occurs in this murine allergen-challenge asthma model. These results suggest that sIL-4R may have beneficial anti-inflammatory effects in asthmatic patients.


Subject(s)
Allergens/administration & dosage , Asthma/pathology , Asthma/prevention & control , Lung/immunology , Lung/pathology , Receptors, Interleukin-4/metabolism , Administration, Intranasal , Animals , Asthma/blood , Asthma/immunology , Bronchial Hyperreactivity/chemically induced , Bronchial Hyperreactivity/immunology , Bronchoalveolar Lavage Fluid/immunology , Cell Movement/immunology , Disease Models, Animal , Eosinophils/pathology , Female , Humans , Immunoglobulin E/blood , Injections, Intraperitoneal , Leukocytes, Mononuclear/pathology , Lung/metabolism , Methacholine Chloride , Mice , Mice, Inbred BALB C , Mucus/immunology , Ovalbumin/administration & dosage , Ovalbumin/immunology , Receptors, Interleukin-4/administration & dosage , Receptors, Interleukin-4/blood , Solubility
10.
Blood ; 95(1): 320-7, 2000 Jan 01.
Article in English | MEDLINE | ID: mdl-10607719

ABSTRACT

Myelokathexis is a congenital disorder that causes severe chronic leukopenia and neutropenia. Characteristic findings include degenerative changes and hypersegmentation of mature neutrophils and hyperplasia of bone marrow myeloid cells. The associated neutropenia can be partially corrected by treatment with granulocyte colony-stimulating factor (G-CSF) or granulocyte-macrophage colony-stimulating factor (GM-CSF). These features led us to propose that accelerated apoptosis of neutrophil precursors might account for the neutropenic phenotype. Blood and bone marrow aspirates were obtained from 4 patients (2 unrelated families) with myelokathexis before G-CSF therapy and from 2 of the affected persons after G-CSF therapy (1 microg/kg per day subcutaneously for 3 weeks). Bone marrow was fractionated using immunomagnetic bead cell sorting into CD34(+), CD33(+)/CD34(-), and CD15(+)/CD34(-)/CD33(- )cell populations. Examination of these cells by flow cytometry and electron microscopy revealed abundant apoptosis in the CD15(+) neutrophil precursor population, characterized by enhanced annexin-V binding, extensive membrane blebbing, condensation of heterochromatin, and cell fragmentation. Colony-forming assays demonstrated significant reduction in a proportion of bone marrow myeloid-committed progenitor cells. Immunohistochemical analysis revealed a selective decrease in bcl-x, but not bcl-2, expression in the CD15(+)/CD34(-)/CD33(-)cell population compared with similar subpopulations of control bone marrow-derived myeloid precursors. After G-CSF therapy, apoptotic features of patients' bone marrow cells were substantially reduced, and the absolute neutrophil counts (ANC) and expression of bcl-x in CD15(+)/CD34(-)/CD33(-)cells increased. The authors concluded that myelokathexis is a disease characterized by the accelerated apoptosis of granulocytes and the depressed expression of bcl-x in bone marrow-derived granulocyte precursor cells. These abnormalities are partially corrected by the in vivo administration of G-CSF. (Blood. 2000;95:320-327)


Subject(s)
Apoptosis , Bone Marrow Cells/pathology , Hematopoietic Stem Cells/pathology , Neutropenia/blood , Neutrophils/physiology , Proto-Oncogene Proteins c-bcl-2/genetics , Adult , Antigens, CD/analysis , Bone Marrow Cells/drug effects , Bone Marrow Cells/ultrastructure , Female , Flow Cytometry , Granulocyte Colony-Stimulating Factor/therapeutic use , Hematopoietic Stem Cells/ultrastructure , Humans , Infant , Male , Neutropenia/congenital , Neutropenia/drug therapy , Neutropenia/pathology , Neutrophils/pathology , Phenotype , Proto-Oncogene Proteins c-bcl-2/analysis , bcl-X Protein
11.
J Immunol ; 163(4): 2217-25, 1999 Aug 15.
Article in English | MEDLINE | ID: mdl-10438964

ABSTRACT

The goals of this study were to determine whether the Fas-dependent apoptosis pathway is active in the lungs of patients with the acute respiratory distress syndrome (ARDS), and whether this pathway can contribute to lung epithelial injury. We found that soluble Fas ligand (sFasL) is present in bronchoalveolar lavage (BAL) fluid of patients before and after the onset of ARDS. The BAL concentration of sFasL at the onset of ARDS was significantly higher in patients who died. BAL from patients with ARDS induced apoptosis of distal lung epithelial cells, which express Fas, and this effect was inhibited by blocking the Fas/FasL system using three different strategies: anti-FasL mAb, anti-Fas mAb, and a Fas-Ig fusion protein. In contrast, BAL from patients at risk for ARDS had no effect on distal lung epithelial cell apoptosis. These data indicate that sFasL is released in the airspaces of patients with acute lung injury and suggest that activation of the Fas/FasL system contributes to the severe epithelial damage that occurs in ARDS. These data provide the first evidence that FasL can be released as a biologically active, death-inducing mediator capable of inducing apoptosis of cells of the distal pulmonary epithelium during acute lung injury.


Subject(s)
Apoptosis/immunology , Epithelial Cells/cytology , Membrane Glycoproteins/physiology , Respiratory Distress Syndrome/immunology , Respiratory Distress Syndrome/pathology , fas Receptor/physiology , Bronchoalveolar Lavage Fluid/immunology , Cell Death/immunology , Cell-Free System/immunology , Cells, Cultured , Epithelial Cells/metabolism , Epithelial Cells/pathology , Fas Ligand Protein , Humans , Ligands , Lung/immunology , Lung/metabolism , Membrane Glycoproteins/metabolism , Pulmonary Alveoli/immunology , Pulmonary Alveoli/metabolism , Respiratory Distress Syndrome/etiology , Respiratory Distress Syndrome/mortality , Risk Factors , Solubility , Survival Analysis
12.
Am J Physiol ; 276(5): L715-27, 1999 05.
Article in English | MEDLINE | ID: mdl-10330027

ABSTRACT

To determine the roles of the type 1 tumor necrosis factor (TNF) receptor (TNFR1) in lung inflammation and antibacterial defense, we exposed transgenic mice lacking TNFR1 [TNFR1(-/-)] and wild-type control mice to aerosolized lipopolysaccharide or Pseudomonas aeruginosa. After LPS, bronchoalveolar lavage fluid (BALF) from TNFR1(-/-) mice contained fewer neutrophils and less macrophage inflammatory protein-2 than BALF from control mice. TNF-alpha, interleukin-1beta, and total protein levels in BALF as well as tissue intercellular adhesion molecule-1 expression did not differ between the two groups. In contrast, lung inflammation and bacterial clearance after infection were augmented in TNFR1(-/-) mice. BALF from infected TNFR1(-/-) mice contained more neutrophils and TNF-alpha and less interleukin-1beta and macrophage inflammatory protein-2 than that from control mice, but protein levels were similarly elevated in both groups. Lung inflammation and bacterial clearance were also augmented in mice lacking both TNF receptors. Thus TNFR1 facilitates neutrophil recruitment after inhalation of lipopolysaccharide, in part by augmenting chemokine induction. In contrast, TNFR1 attenuates lung inflammation in response to live bacteria but does not contribute to increased lung permeability and is not required for the elimination of P. aeruginosa.


Subject(s)
Endotoxins/administration & dosage , Pneumonia, Bacterial/microbiology , Pseudomonas Infections , Receptors, Tumor Necrosis Factor/physiology , Administration, Inhalation , Animals , Bronchoalveolar Lavage Fluid/chemistry , Bronchoalveolar Lavage Fluid/cytology , Chemokine CXCL2 , Chemotaxis, Leukocyte , Cytokines/analysis , Intercellular Adhesion Molecule-1/analysis , Interleukin-1/analysis , Leukocyte Count , Mice , Mice, Inbred C57BL , Mice, Transgenic , Monokines/analysis , Neutrophils , Pneumonia, Bacterial/metabolism , Pneumonia, Bacterial/pathology , Proteins/analysis , Receptors, Tumor Necrosis Factor/deficiency , Receptors, Tumor Necrosis Factor/genetics , Tumor Necrosis Factor-alpha/analysis
13.
J Thorac Cardiovasc Surg ; 117(5): 931-8, 1999 May.
Article in English | MEDLINE | ID: mdl-10220688

ABSTRACT

INTRODUCTION: Pulmonary arteriovenous malformations are a common cause of progressive cyanosis in children after cavopulmonary anastomoses. We analyzed the pulmonary histologic characteristics from children in whom pulmonary arteriovenous malformations developed after procedures that resulted in pulmonary arterial blood flow devoid of hepatic venous effluent. METHODS: We performed routine histologic studies, immunohistochemical staining, and electron microscopic analysis of peripheral lung biopsy specimens from 2 children with angiographically proven pulmonary arteriovenous malformations. Microvessel density was determined with a computer-assisted, morphometric analysis system. RESULTS: Histologic examination demonstrated large, dilated blood vessels ("lakes") and clustered, smaller vessels ("chains") in the pulmonary parenchyma. Microvessel density was significantly greater in these patients than in age-matched controls (P =.01). Immunohistochemistry demonstrated uniform staining for type IV collagen and alpha-smooth muscle actin, weak staining for the endothelial marker CD31 (cluster of differentiation, PECAM-1), and negative staining for proliferating cell nuclear antigen. Electron microscopy revealed endothelial irregularity, a disorganized basement membrane, and increased numbers of collagen and actin filaments beneath the endothelium. CONCLUSIONS: This study represents an attempt to characterize the histologic features of pulmonary arteriovenous malformations in children with congenital heart disease who have pulmonary arterial blood flow devoid of hepatic venous effluent. The histologic correlate of this condition appears to be greatly increased numbers of thin-walled vessels. Immunohistochemistry suggests that the rate of cellular proliferation is not increased in these lesions. The development of these techniques may provide a standardized histologic approach for this condition and aid in understanding its etiology.


Subject(s)
Arteriovenous Malformations/pathology , Cyanosis/complications , Heart Defects, Congenital/complications , Pulmonary Artery/abnormalities , Pulmonary Veins/abnormalities , Anastomosis, Surgical/adverse effects , Angiography , Arteriovenous Malformations/diagnostic imaging , Arteriovenous Malformations/etiology , Biopsy , Capillaries/diagnostic imaging , Capillaries/ultrastructure , Child , Child, Preschool , Cyanosis/surgery , Female , Follow-Up Studies , Heart Atria/surgery , Heart Defects, Congenital/surgery , Hepatic Veins/surgery , Humans , Lung/blood supply , Lung/ultrastructure , Male , Pulmonary Artery/pathology , Pulmonary Veins/pathology , Vena Cava, Superior/surgery
14.
Am J Respir Cell Mol Biol ; 19(6): 881-91, 1998 Dec.
Article in English | MEDLINE | ID: mdl-9843922

ABSTRACT

The pulmonary host response to infection and inflammation appears, at least in part, to be compartmentalized from the systemic host response. Tumor necrosis factor-alpha (TNF-alpha) has been implicated in lung inflammation and injury, but its site(s) of action has not been clearly defined. To investigate this, transgenic mice (surfactant apoprotein C promotor/soluble TNF receptor type II-Fc fusion protein ([SPCTNFRIIFc] mice) were generated in which TNF-alpha was selectively antagonized in the distal lung through tissue-specific expression of sTNFRIIFc, a soluble TNF inhibitor. The lung inflammatory response in these mice to pulmonary challenge with Micropolyspora faeni antigen or lipopolysaccharide (LPS) was compared with the response of wild-type mice, wild-type mice treated with recombinant sTNFRIIFc intravenously, and type I TNF-receptor knockout mice. Recruitment of polymorphonuclear leukocytes (PMN) to the lung after challenge with M. faeni antigen was essentially abolished in the TNFRI knockout mice and markedly reduced in the SPCTNFRIIFc mice. Wild-type mice given sTNFRIIFc intravenously in amounts resulting in lung concentrations similar to those in SPCTNFRIIFc mice also showed significantly reduced lung PMN recruitment, whereas those given doses that achieved such concentrations in the blood but low levels in the lung did not. In contrast, PMN recruitment to the lung following aerosol challenge with LPS was reduced significantly in the TNFRI knockout mice and in mice given high-dose sTNFRIIFc intravenously, but was not reduced significantly in SPCTNFRIIFc mice. Thus, inhibition of PMN recruitment in response to M. faeni antigen correlated largely with the extent of intrapulmonary inhibition of TNF-alpha, whereas the response to LPS correlated best with the extent of extrapulmonary inhibition of TNF-alpha. These studies indicate that TNF-alpha may act at different loci to mediate lung inflammation, with the site of action depending in part on the nature of the inflammatory stimulus, and that SPCTNFRIIFc mice provide a tool by which the locus of TNF action can be addressed.


Subject(s)
Pneumonia/immunology , Tumor Necrosis Factor-alpha/physiology , Administration, Intranasal , Animals , Antigens, Bacterial/administration & dosage , Antigens, Bacterial/immunology , Apolipoproteins C/genetics , Immunoglobulin Fc Fragments/genetics , Lipopolysaccharides/administration & dosage , Lung/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Micromonosporaceae/immunology , Neutrophils/pathology , Pneumonia/pathology , Promoter Regions, Genetic , Receptors, Tumor Necrosis Factor/genetics , Recombinant Fusion Proteins/pharmacology
15.
Ultrasound Med Biol ; 24(6): 903-10, 1998 Jul.
Article in English | MEDLINE | ID: mdl-9740391

ABSTRACT

The hemorrhagic complications of vascular injury can be significant. We report on the use of high-intensity focused ultrasound (HIFU) to stop the hemorrhage of punctured blood vessels in pigs. Two HIFU transducers with frequencies of 3.5 and 2.0 MHz, each equipped with a water-filled conical housing, were used. Major blood vessels (femoral artery and vein, axillary artery, carotid artery and jugular vein), 2-10 mm in diameter, of anesthetized pigs were exposed surgically and punctured with 14- and 18-gauge needles to produce moderate to profuse bleeding. Complete hemostasis was achieved in less than 3 min of HIFU treatment in most blood vessels, and all vessels were patent after the treatment. Both HIFU frequencies were effective in producing hemostasis. Gross examination of the HIFU-treated vessels showed a consistent hardening of the soft tissue surrounding the blood vessels, providing a seal for the puncture hole. Microscopic examination of the vessels showed a remarkably localized HIFU treatment, resulting in coagulation of the adventitia, and an extensive fibrin network around the vessels and in the puncture hole. The vessel walls exhibited focal swelling, without evidence of irreversible injury. HIFU may provide a useful method for achieving hemostasis of punctured and traumatized blood vessels in a variety of clinical settings.


Subject(s)
Blood Vessels/diagnostic imaging , Hemorrhage/therapy , Hemostatic Techniques , Ultrasonic Therapy , Animals , Blood Vessels/injuries , Female , Hemorrhage/diagnostic imaging , Male , Swine , Ultrasonography
16.
J Infect Dis ; 177(5): 1437-43, 1998 May.
Article in English | MEDLINE | ID: mdl-9593043

ABSTRACT

Mast cells participate in the host defense against parasites. Mast cells release leukotrienes (LTs), potent 5-lipoxygenase (LO) products of arachidonic acid well-known to be involved in the inflammatory process. After incubation with Toxoplasma gondii, mast cells were found to degranulate and release LTB4; this interaction damages the tachyzoites. This mast cell activity against the tachyzoites was inhibited by the 5-LO inhibitor A-63162 and the 5-LO-activating protein inhibitor MK-886 but not by the cyclooxygenase inhibitor indomethacin. Reactive oxygen species were not implicated in the mast cell-mediated toxoplasmacidal activity. The generation of LTs is important for mast cell secretion, and LTB4 released by mast cells and other inflammatory cells may be a key factor in the host defense against T. gondii.


Subject(s)
Arachidonate 5-Lipoxygenase/metabolism , Leukotriene B4/biosynthesis , Leukotrienes/physiology , Mast Cells/physiology , Mast Cells/parasitology , Toxoplasma/physiology , Acetamides/pharmacology , Animals , Cell Adhesion , Cells, Cultured , Enzyme Activation , Indoles/pharmacology , Leukotrienes/biosynthesis , Lipoxygenase Inhibitors/pharmacology , Male , Mast Cells/ultrastructure , Microscopy, Electron , Phenyl Ethers , Rats , Rats, Sprague-Dawley , Toxoplasma/drug effects , Toxoplasma/ultrastructure
17.
Infect Immun ; 65(12): 5074-81, 1997 Dec.
Article in English | MEDLINE | ID: mdl-9393798

ABSTRACT

Group B streptococci (GBS) are the leading cause of meningitis in newborns. Although meningitis develops following bacteremia, the precise mechanism or mechanisms whereby GBS leave the bloodstream and gain access to the central nervous system (CNS) are not known. We hypothesized that GBS produce meningitis because of a unique capacity to invade human brain microvascular endothelial cells (BMEC), the single-cell layer which constitutes the blood-brain barrier. In order to test this hypothesis, we developed an in vitro model with BMEC isolated from a human, immortalized by simian virus 40 transformation, and propagated in tissue culture monolayers. GBS invasion of BMEC monolayers was demonstrated by electron microscopy. Intracellular GBS were found within membrane-bound vacuoles, suggesting the organism induced its own endocytic uptake. GBS invasion of BMEC was quantified with a gentamicin protection assay. Serotype III strains, which account for the majority of CNS isolates, invaded BMEC more efficiently than strains from other common GBS serotypes. GBS survived within BMEC for up to 20 h without significant intracellular replication. GBS invasion of BMEC required active bacterial DNA, RNA, and protein synthesis, as well as microfilament and microtubule elements of the eukaryotic cytoskeleton. The polysaccharide capsule of GBS attenuated the invasive ability of the organism. At high bacterial densities, GBS invasion of BMEC was accompanied by evidence of cellular injury; this cytotoxicity was correlated to beta-hemolysin production by the bacterium. Finally, GBS demonstrated transcytosis across intact, polar BMEC monolayers grown on Transwell membranes. GBS invasion of BMEC may be a primary step in the pathogenesis of meningitis, allowing bacteria access to the CNS by transcytosis or by injury and disruption of the endothelial blood-brain barrier.


Subject(s)
Endothelium, Vascular/microbiology , Microcirculation/microbiology , Streptococcus/physiology , Brain/blood supply , Cells, Cultured , Endothelium, Vascular/ultrastructure , Humans , Microscopy, Electron , Streptococcus/pathogenicity
18.
Am J Respir Crit Care Med ; 156(6): 1969-77, 1997 Dec.
Article in English | MEDLINE | ID: mdl-9412582

ABSTRACT

Little is known about neutrophil (PMN) apoptosis in the acute respiratory distress syndrome (ARDS). We uses morphologic criteria to count apoptotic PMN in bronchoalveolar lavage fluid (BAL) of 35 patients on Days 1, 3, 7, 14, and 21 of ARDS and 13 patients on Days 1 and 3 of risk for ARDS. We found that the proportion of apoptotic PMN in BAL was low throughout the course of ARDS. There was no significant difference between the percentage of apoptotic PMN in patients at risk and patients with established ARDS or between patients who lived (2.4%) and patients who died (1.8%). When normal human PMN were incubated in ARDS BAL, a significantly lower proportion became apoptotic (50 +/- 4%), as compared with PMN incubated in lavage fluid from normal volunteers (76 +/- 7%, p < 0.05). This antiapoptotic effect of ARDS BAL was blocked by immunodepleting BAL of G-CSF and GM-CSF. We conclude that the proportion of apoptotic PMN recovered from the lungs of patients with ARDS is low throughout the course of ARD S. Furthermore, BAL from patients with ARDS prolongs survival of normal human PMN in vitro, and this effect is partially mediated by G-CSF and GM-CSF.


Subject(s)
Apoptosis , Neutrophils/pathology , Respiratory Distress Syndrome/pathology , Adult , Aged , Bronchoalveolar Lavage Fluid/chemistry , Bronchoalveolar Lavage Fluid/cytology , Cell Survival , Female , Granulocyte Colony-Stimulating Factor/analysis , Granulocyte-Macrophage Colony-Stimulating Factor/analysis , Humans , In Vitro Techniques , Interferon-gamma/analysis , Interleukin-6/analysis , Leukocyte Count , Male , Middle Aged , Respiratory Distress Syndrome/metabolism , Risk Factors
19.
Am J Respir Crit Care Med ; 155(6): 2030-40, 1997 Jun.
Article in English | MEDLINE | ID: mdl-9196112

ABSTRACT

The major goals of this study were to define the relationships between intrapulmonary and systemic inflammatory responses in animals with gram-negative pneumonia. We treated rabbits with intrapulmonary Escherichia coli (1 x 10(7) to 1 x 10(10) cfu/ml), and then measured physiologic, cellular, and molecular events in the lungs and systemic circulation for 24 h. The treatment protocols resulted in groups of animals that mimicked the stages of the septic inflammatory response in humans. Animals treated with low inocula had systemic changes consistent with systemic inflammatory response syndrome and cleared the bacteria and inflammatory products from the lungs. Animals treated with high inocula failed to clear bacteria from the lungs, had severe intrapulmonary inflammatory responses, and developed septic shock. Intrapulmonary leukocyte recruitment was directly related to the size of the bacterial inoculum, but lung protein accumulation was not. Tumor neurosis factor-alpha (TNF-alpha), interleukin-8 (IL-8), and GRO were detectable in lung lavage fluid at 4 h and declined by 24 h in animals that cleared intrapulmonary E. coli. In contrast, lavage TNF-alpha, IL-8, and GRO increased over 24 h in animals that failed to clear intrapulmonary bacteria. MCP-1 increased between 4 h and 24 h in the lungs of all of the animals as the histologic response evolved from neutrophilic to mononuclear cell predominance. Thus, the intensity of systemic inflammatory and physiologic responses to intrapulmonary gram-negative infection depends on the inoculum size and whether the bacteria are cleared from or proliferate in the lungs. The results provide experimental support for the recently proposed classification of septic responses in humans.


Subject(s)
Escherichia coli Infections , Inflammation/microbiology , Pneumonia/microbiology , Animals , Blood/microbiology , Bronchoalveolar Lavage Fluid/chemistry , Culture Techniques , Cytokines/analysis , Cytokines/blood , Endotoxins/blood , Escherichia coli/isolation & purification , Lung/metabolism , Lung/microbiology , Lung/pathology , Pneumonia/metabolism , Pneumonia/pathology , Rabbits
20.
Am J Respir Crit Care Med ; 155(2): 661-9, 1997 Feb.
Article in English | MEDLINE | ID: mdl-9032210

ABSTRACT

We used various ovalbumin sensitization and challenge protocols to determine the importance of the route of allergen administration and the genetic background in modulating the physiologic, inflammatory, and immunologic features characteristic of allergen-induced asthma. In BALB/c mice, induction of maximal airway hyperresponsiveness and airspace eosinophilia required administration of ovalbumin by both the intraperitoneal and the intranasal routes (combination protocol), whereas intraperitoneal immunization alone resulted in maximal ovalbumin-specific IgE plasma levels. Thus, a systemic immune response to allergen, in addition to, or independent of IgE production, as well as local allergen challenge were necessary for maximal induction of pulmonary disease. BALB/c mice treated with ovalbumin by the combination protocol had increased Th2-type cytokine mRNA levels in bronchial lymph node tissue compared with control mice. In contrast, C57BL/6 mice treated with ovalbumin by the combination protocol had significantly decreased responses compared with BALB/c mice for all parameters of allergic pulmonary disease examined, with the exception of airspace eosinophilia. Genetic background has a striking and selective effect on the phenotype of murine allergic pulmonary disease. Further analysis of this murine model should be useful in helping define the critical pathogenetic events in allergen-induced asthma.


Subject(s)
Allergens/genetics , Lung Diseases/etiology , Lung Diseases/genetics , Lung Diseases/immunology , Ovalbumin/administration & dosage , Serine Proteinase Inhibitors/administration & dosage , Administration, Intranasal , Animals , Antibody Formation , Bronchi/pathology , Bronchoalveolar Lavage Fluid/chemistry , Female , Immunoglobulin E/blood , Injections, Intraperitoneal , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Phenotype , Species Specificity
SELECTION OF CITATIONS
SEARCH DETAIL
...