Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 71
Filter
1.
Regul Toxicol Pharmacol ; 59(3): 445-53, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21295096

ABSTRACT

A Type III Built-up Roofing Asphalt (BURA) fume condensate was evaluated for subchronic systemic toxicity and reproductive/developmental toxicity screening in Wistar rats, by OECD protocol 422 and OECD cytogenetic protocol 474. Animals were exposed by nose-only inhalation to target concentrations of 30, 100 and 300 mg/m³ total hydrocarbons (actual concentrations, 30.0, 100.1 and 297.3 mg/m³). The study was performed to assess potential hazards from asphalt fumes to which humans could be exposed during application. No adverse effects were seen for spermology, reproductive or developmental parameters or early postnatal development of offspring from day 1 to 4 postpartum. BURA fume condensate did not induce any significant increases in micronucleus frequency in polychromatic erythrocytes of rat bone marrow nor was neurobehavioral toxicity observed at any dose. Systemic effects were slight and seen at doses above those measured at work sites. The systemic NOAEC of 100 mg/m³ for males was based on decreased body weight gain, food consumption and increased absolute and relative lung wet weight correlated with slight histological changes in the lung, primarily adaptive in nature at 300 mg/m³. The female NOAEC of 30 mg/m³ was based on a statistically significant increase in relative wet lung weight at higher doses, correlated with slight histopathologic effects in the lungs at the highest dose. However, no increase in relative lung weight was seen in breeding females at 100 mg/m³.


Subject(s)
Hydrocarbons/administration & dosage , Hydrocarbons/toxicity , Inhalation Exposure , Lung/drug effects , Reproduction/drug effects , Administration, Inhalation , Administration, Intranasal , Animals , Body Weight/drug effects , Body Weight/physiology , Cytogenetic Analysis/methods , Drug Administration Schedule , Drug Evaluation, Preclinical/methods , Environmental Monitoring/methods , Female , Inhalation Exposure/adverse effects , Lung/growth & development , Male , Pregnancy , Rats , Rats, Wistar , Reproduction/physiology
2.
Thorax ; 61(4): 354-61, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16565268

ABSTRACT

Exacerbations of chronic obstructive pulmonary disease (COPD) are associated with increased morbidity and mortality. The effective management of COPD exacerbations awaits a better understanding of the underlying pathophysiological mechanisms that shape its clinical expression. The clinical presentation of exacerbations of COPD is highly variable and ranges from episodic symptomatic deterioration that is poorly responsive to usual treatment, to devastating life threatening events. This underscores the heterogeneous physiological mechanisms of this complex disease, as well as the variation in response to the provoking stimulus. The derangements in ventilatory mechanics, muscle function, and gas exchange that characterise severe COPD exacerbations with respiratory failure are now well understood. Critical expiratory flow limitation and the consequent dynamic lung hyperinflation appear to be the proximate deleterious events. Similar basic mechanisms probably explain the clinical manifestations of less severe exacerbations of COPD, but this needs further scientific validation. In this review we summarise what we have learned about the natural history of COPD exacerbations from clinical studies that have incorporated physiological measurements. We discuss the pathophysiology of clinically stable COPD and examine the impact of acutely increased expiratory flow limitation on the compromised respiratory system. Finally, we review the chain of physiological events that leads to acute ventilatory insufficiency in severe exacerbations.


Subject(s)
Pulmonary Disease, Chronic Obstructive/physiopathology , Acute Disease , Heart Diseases/etiology , Humans , Prognosis , Pulmonary Disease, Chronic Obstructive/etiology , Pulmonary Disease, Chronic Obstructive/therapy , Respiration, Artificial , Respiratory Function Tests , Respiratory Insufficiency/etiology
3.
Eur Respir J ; 26(3): 420-8, 2005 Sep.
Article in English | MEDLINE | ID: mdl-16135722

ABSTRACT

Acute exacerbations of chronic obstructive disease (AECOPD) are characterised by worsening dyspnoea that is variably prolonged. In this study, physiological changes during moderate AECOPD were examined and the factors associated with dyspnoea resolution over time were determined. In total, 20 patients experiencing an AECOPD were evaluated within 72 h of initial worsening of symptoms (day 0) with pulmonary function testing, metabolic testing and symptom assessment using the dyspnoea domain of the Chronic Respiratory Disease Questionnaire (CRQ). Treatment was optimised and testing was repeated after 7, 14, 30 and 60 days. At day 0, patients were very short of breath (CRQ-dyspnoea mean+/-SEM 2.4+/-0.3) and showed significant airflow obstruction (forced expiratory volume in one second (FEV1) 41+/-3% predicted) and lung hyperinflation (forced residual capacity (FRC) 164+/-7% pred). By day 60 CRQ-dyspnoea improved to 4.6+/-0.5 (some shortness of breath); FRC and residual volume decreased by 5 and 11%, respectively; inspiratory capacity (IC) and slow vital capacity increased by 18 and 17%, respectively; and FEV1 increased by 18% with no change in FEV1/FVC. Total lung capacity did not change during AECOPD, and thus, changes in IC reliably reflected changes in end-expiratory lung volume. In conclusion, moderate acute exacerbation of chronic obstructive pulmonary disease is characterised by worsening airflow obstruction and lung hyperinflation. Improvement of dyspnoea following acute exacerbations of chronic obstructive pulmonary disease was associated with reduction in lung hyperinflation and consequent increase in expiratory flow rates.


Subject(s)
Dyspnea/etiology , Dyspnea/physiopathology , Lung/physiopathology , Pulmonary Disease, Chronic Obstructive/complications , Pulmonary Disease, Chronic Obstructive/physiopathology , Recovery of Function/physiology , Acute Disease , Aged , Female , Health Surveys , Humans , Male , Middle Aged , Prospective Studies , Reproducibility of Results , Respiratory Function Tests , Severity of Illness Index , Time Factors
4.
J Exp Med ; 194(9): 1243-52, 2001 Nov 05.
Article in English | MEDLINE | ID: mdl-11696590

ABSTRACT

Mast cells (MCs) are centrally important in allergic inflammation of the airways, as well as in the intestinal immune response to helminth infection. A single lineage of bone marrow (BM)-derived progenitors emigrates from the circulation and matures into phenotypically distinct MCs in different tissues. Because the mechanisms of MC progenitor (MCp) homing to peripheral tissues have not been evaluated, we used limiting dilution analysis to measure the concentration of MCp in various tissues of mice deficient for candidate homing molecules. MCp were almost completely absent in the small intestine but were present in the lung, spleen, BM, and large intestine of beta7 integrin-deficient mice (on the C57BL/6 background), indicating that a beta7 integrin is critical for homing of these cells to the small intestine. MCp concentrations were not altered in the tissues of mice deficient in the alphaE integrin (CD103), the beta2 integrin (CD18), or the recombination activating gene (RAG)-2 gene either alone or in combination with the interleukin (IL)-receptor common gamma chain. Therefore, it is the alpha4beta7 integrin and not the alphaEbeta7 integrin that is critical, and lymphocytes and natural killer cells play no role in directing MCp migration under basal conditions. When MCp in BALB/c mice were eliminated with sublethal doses of gamma-radiation and then reconstituted with syngeneic BM, the administration of anti-alpha4beta7 integrin, anti-alpha4 integrin, anti-beta7 integrin, or anti-MAdCAM-1 monoclonal antibodies (mAbs) blocked the recovery of MCp in the small intestine. The blocking mAbs could be administered as late as 4 d after BM reconstitution with optimal inhibition, implying that the MCp must arise first in the BM, circulate in the vasculature, and then translocate into the intestine. Inasmuch as MCp are preserved in the lungs of beta7 integrin-deficient and anti-alpha4beta7 integrin-treated mice but not in the small intestine, alpha4beta7 integrin is critical for tissue specific extravasation for localization of MCp in the small intestine, but not the lungs.


Subject(s)
Antigens, CD/immunology , Chemotaxis/immunology , Integrin alpha Chains , Integrin beta Chains , Integrins/immunology , Intestines/immunology , Mast Cells/immunology , Stem Cells/immunology , Animals , Antibodies, Monoclonal/immunology , CD18 Antigens/immunology , Cell Count , Cytokines/administration & dosage , Cytokines/immunology , Integrin alpha4 , Integrins/genetics , Intestines/cytology , Mast Cells/cytology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Proto-Oncogene Proteins c-kit/immunology , Stem Cell Factor/immunology , Stem Cells/cytology
5.
J Invest Dermatol ; 117(3): 569-75, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11564161

ABSTRACT

Tissue-specific T cell localization is crucial for immune surveillance of normal tissues and the pathogenesis of inflammatory disorders. In psoriatic skin, CD8+ lymphocytes predominantly reside within the epidermis, whereas CD4+ T cells are most abundant within the dermis. Molecular mechanisms guiding this spatial compartmentalization are not completely understood, however. Here, we demonstrate that 55% (+/-9.7%, n = 14) of the epidermal T cells, predominantly of the CD8+ phenotype, expressed the integrin alphaE(CD103)beta7. In contrast, only 5% (+/-2.0%) of the dermal T cells were alphaE(CD103)beta7+. Integrin alphaE(CD103)beta7 was not detected in normal skin (n = 10), and less than 1% of peripheral blood lymphocytes derived from normal (n = 11) or psoriatic (n = 10) donors expressed alphaE(CD103). When cultured T lymphoblasts (n = 12 donors) were stimulated with transforming growth factor beta1, expression of integrin alphaE(CD103)beta7 was induced on 52.8% (+/-16.2%) of CD8+ cells, but only on 6.1% (+/-2.3%) of CD4+ cells, suggesting selective inducibility on CD8+ lymphocytes. Whereas similar overall expression of transforming-growth-factor-beta1-specific mRNA was detected in normal and psoriatic skin by real-time quantitative polymerase chain reaction, immunohistochemistry revealed focal overexpression of transforming growth factor beta1 underneath psoriatic, but not normal, epidermis. This heterogenous transforming growth factor beta1 expression may contribute to induction of alphaE(CD103) in vivo. Adhesion of transforming-growth-factor-beta1-stimulated CD8+, but not CD4+, T cells to cultured keratinocytes and psoriatic epidermis in frozen sections could be significantly inhibited by antibodies that blocked the alphaE(CD103)/E-cadherin interaction. Co-culture of lymphoblasts and keratinocytes resulted in marginal enhancement of alphaE(CD103)beta7 expression in some cases. Overall, integrin alphaE(CD103)beta7 appears to contribute to tissue-specific epidermal localization of CD8+ T lymphocytes.


Subject(s)
Antigens, CD/immunology , CD8-Positive T-Lymphocytes/immunology , Epidermis/immunology , Integrin alpha Chains , CD8-Positive T-Lymphocytes/cytology , Cells, Cultured , Cytotoxicity, Immunologic , Epidermal Cells , Humans , Psoriasis/immunology , Psoriasis/pathology
6.
J Immunol ; 166(5): 3506-14, 2001 Mar 01.
Article in English | MEDLINE | ID: mdl-11207310

ABSTRACT

Integrins are important for T cell interactions with endothelial cells. Because the integrin alpha(E)beta(7) is expressed on some circulating gut-homing T cells and as T cell numbers are reduced in the intestinal lamina propria of alpha(E)-deficient mice, we evaluated whether alpha(E)beta(7) mediates binding to intestinal endothelial cells. We found that anti-alpha(E)beta(7) mAbs partially blocked the binding of cultured intraepithelial T cells to human intestinal microvascular endothelial cells (HIMEC). Furthermore, alpha(E)beta(7)-transfected K562 cells bound more efficiently than vector-transfected K562 cells to HIMEC. Finally, HIMEC bound directly to an alpha(E)beta(7)-Fc fusion protein. These interactions were partially blocked by anti-alpha(E)beta(7) mAbs, and endothelial cell binding to the alpha(E)beta(7)-Fc was dependent upon the metal ion-dependent adhesion site within the alpha(E) A domain. Of note, the HIMEC lacked expression of E-cadherin, the only known alpha(E)beta(7) counterreceptor as assessed by functional studies, flow cytometry, and RT-PCR. Thus, HIMEC/alpha(E)beta(7) binding was independent of E-cadherin. In addition, this interaction appeared to be tissue selective, as HIMEC bound to the alpha(E)beta(7)-Fc, whereas microvascular endothelial cells from the skin did not. Finally, there was evidence for an alpha(E)beta(7) ligand on intestinal endothelial cells in vivo, as alpha(E)beta(7) expression enhanced lymphocyte binding around vessels in the lamina propria in tissue sections. Thus, we have defined a novel interaction for alpha(E)beta(7) at a nonepithelial location. These studies suggest a role for alpha(E)beta(7) in interactions with the intestinal endothelium that may have implications for intestinal T cell homing or functional responses.


Subject(s)
Antigens, CD/physiology , Cadherins/physiology , Endothelium, Vascular/immunology , Integrin alpha Chains , Integrins/physiology , Intestinal Mucosa/blood supply , Intestinal Mucosa/immunology , Antibodies, Blocking/metabolism , Antibodies, Blocking/pharmacology , Antibodies, Monoclonal/metabolism , Antibodies, Monoclonal/pharmacology , Antigens, CD/biosynthesis , Antigens, CD/genetics , Antigens, CD/immunology , Binding Sites, Antibody , Binding, Competitive/immunology , Cell Adhesion/genetics , Cell Adhesion/immunology , Cell Communication/immunology , Cell Line , Cell Line, Transformed , Cells, Cultured , Endothelium, Vascular/cytology , Endothelium, Vascular/metabolism , Genetic Vectors/biosynthesis , Genetic Vectors/metabolism , Humans , Immunoglobulin Fc Fragments/genetics , Immunoglobulin Fc Fragments/metabolism , Integrins/biosynthesis , Integrins/genetics , Integrins/immunology , Intestinal Mucosa/cytology , Intestinal Mucosa/metabolism , K562 Cells , Metals/metabolism , Microcirculation/cytology , Microcirculation/immunology , Microcirculation/metabolism , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/immunology , Recombinant Fusion Proteins/metabolism , Skin/blood supply , Skin/cytology , Skin/immunology , Skin/metabolism , Solubility , Tumor Cells, Cultured
7.
J Immunol ; 165(11): 6583-9, 2000 Dec 01.
Article in English | MEDLINE | ID: mdl-11086102

ABSTRACT

The integrin alpha(E)beta(7) is thought to play an important role in the localization of mucosal, but not of cutaneous T lymphocytes. Thus, it was surprising that 89% of adult alpha(E)(-/-) mice on the 129/Sv x BALB/c background developed inflammatory skin lesions without an apparent infectious etiology. Skin inflammation correlated with alpha(E) deficiency in mice with a mixed 129/Sv x BALB/c background, but not in mice further backcrossed to BALB/c and housed in a second animal facility. These studies suggested that alpha(E) deficiency, in combination with other genetic and/or environmental factors, is involved in lesion development. The lesions were infiltrated by CD4(+) T cells and neutrophils, and associated with increased expression of inflammatory cytokines. Furthermore, skin inflammation resulted from transfer of unfractionated alpha(E)(-/-) splenocytes into scid/scid mice, but not from transfer of wild-type splenocytes, suggesting that the lesions resulted from immune dysregulation. We also studied the role of alpha(E)beta(7) in a murine model of hyperproliferative inflammatory skin disorders that is induced by transfer of minor histocompatibility-mismatched CD4(+)/CD45RB(high) T cells into scid/scid mice under specific environmental conditions. Under housing conditions that were permissive for lesion development, transfer of alpha(E)-deficient CD4(+)/CD45RB(high) T cells significantly exacerbated the cutaneous lesions as compared with lesions observed in mice reconstituted with wild-type donor cells. These experiments suggested that alpha(E)-expressing cells play an important role during the course of cutaneous inflammation. In addition, they suggest that alpha(E)beta(7) deficiency, in combination with other genetic or environmental factors, is a risk factor for inflammatory skin disease.


Subject(s)
Antigens, CD/genetics , Integrin alpha Chains , Integrins/deficiency , Integrins/genetics , Skin/pathology , Adoptive Transfer , Aging/genetics , Aging/immunology , Animals , Autoimmune Diseases/genetics , Autoimmune Diseases/immunology , Autoimmune Diseases/pathology , Cell Division/genetics , Cell Division/immunology , Disease Models, Animal , Inflammation/genetics , Inflammation/immunology , Integrins/physiology , Mice , Mice, Knockout , Mice, SCID , Psoriasis/genetics , Psoriasis/immunology , Psoriasis/pathology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , T-Lymphocyte Subsets/transplantation
8.
J Immunol ; 165(8): 4667-75, 2000 Oct 15.
Article in English | MEDLINE | ID: mdl-11035110

ABSTRACT

Mice chimeric for integrins alpha(5), alpha(V), or alpha(4) were used to dissect the in vivo roles of these adhesion receptors during leukocyte development and traffic. No major defects were observed in the development of lymphocytes, monocytes, or granulocytes or in the traffic of lymphocytes to different lymphoid organs in the absence of alpha(5) or alpha(V) integrins. However, in agreement with previous reports, the absence of alpha(4) integrins produced major defects in development of lymphoid and myeloid lineages and a specific defect in homing of lymphocytes to Peyer's patches. In contrast, the alpha(4) integrin subunit is not essential for localization of T lymphocytes into intraepithelial and lamina propria compartments in the gut, whereas one of the partners of alpha(4), the beta(7) chain, has been shown to be essential. However, alpha(4)-deficient T lymphocytes cannot migrate properly during the inflammatory response induced by thioglycolate injection into the peritoneum. Finally, in vitro proliferation and activation of lymphocytes deficient for alpha(5), alpha(V), or alpha(4) integrins upon stimulation with different stimuli were similar to those seen in controls. These results show that integrins play distinct roles during in vivo leukocyte development and traffic.


Subject(s)
Cell Movement/immunology , Chimera/immunology , Integrins/physiology , Leukocytes/cytology , Leukocytes/immunology , Animals , Antigens, CD/genetics , Antigens, CD/physiology , Cell Division/genetics , Cell Division/immunology , Cell Movement/genetics , Integrin alpha4 , Integrin alpha5 , Integrin alphaV , Integrins/genetics , Lymphocyte Activation/genetics , Lymphocytes/cytology , Lymphocytes/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Myeloid Cells/immunology , Peritonitis/chemically induced , Peritonitis/genetics , Peritonitis/immunology , Receptors, Lymphocyte Homing/physiology , Thioglycolates/administration & dosage
9.
Reg Anesth Pain Med ; 25(5): 506-13, 2000.
Article in English | MEDLINE | ID: mdl-11009237

ABSTRACT

BACKGROUND AND OBJECTIVES: The purpose of this study was to determine whether the addition of ketorolac tromethamine to local anesthesia for ankle block alters the quality or duration of analgesia after podiatric surgery. The second aim was to determine the chemical stability of ketorolac tromethamine when added to local anesthetic solutions. METHODS: The study design was double-blinded, placebo-controlled, and randomized. Seventy-nine American Society of Anesthesiologists (ASA) class I or II patients scheduled for bunionectomy or hammer toe repair, or both were randomized to 1 of 4 groups. Group L received plain 1.73% lidocaine for their ankle block. Group K received 1.73% lidocaine with ketorolac (4 mg/mL) added to the local solution. Group Kiv received 1.73% plain lidocaine for ankle block and 20 mg of ketorolac intravenously. Group E received 1.73% lidocaine with .67% ethanol added. The final concentration of lidocaine for all groups was 1.73%. The block performed in each patient was a 5-point ankle block. Beginning at 1 hour after the completion of the block and every 30 minutes thereafter, visual analogue scale (VAS) and verbal pain scores were recorded. The time from performance of the block to the initial pain and time to the first oral pain medication intake were also recorded. The time and amount of postoperative oral analgesics in the first 9 hours after the block were recorded. Adverse events were also recorded for each group. RESULTS: There were significantly lower overall VAS and verbal pain scores for group K compared with groups E and L and group Kiv compared with group E. Group K also had a significantly longer time to the first reported pain and first oral pain medications than groups E and L, but not with Group Kiv. The same group had significantly fewer average doses of pain medications postoperatively than Groups E and L. Group E had significantly shorter times to first report of pain and first pain medications and higher mean dose of postoperative oral analgesics than group K and Group Kiv. There were no untoward side effects reported from any group. Chemical analysis by gas chromatography (GC) and capillary electrophoresis (CE) showed no significant change in composition of the solutions when ketorolac was mixed with lidocaine and/or bupivacaine and stored at 37 degrees C for 1 week. CONCLUSIONS: The addition of ketorolac to lidocaine for ankle block contributed to longer duration and better quality analgesia after foot surgery compared with plain 1.73% lidocaine or 1.73% lidocaine plus intravenous ketorolac. The ethanol vehicle is unlikely responsible for the analgesic effects of ketorolac. Ketorolac retains its chemical stability when placed in local solutions of lidocaine or bupivacaine.


Subject(s)
Anesthetics, Local/administration & dosage , Anti-Inflammatory Agents, Non-Steroidal/administration & dosage , Hallux Valgus/surgery , Ketorolac/administration & dosage , Lidocaine/therapeutic use , Nerve Block , Pain, Postoperative/drug therapy , Adult , Aged , Double-Blind Method , Drug Stability , Electrophoresis, Capillary , Female , Humans , Lidocaine/administration & dosage , Lidocaine/chemistry , Male , Middle Aged , Prospective Studies
10.
Curr Opin Cell Biol ; 12(5): 563-8, 2000 Oct.
Article in English | MEDLINE | ID: mdl-10978890

ABSTRACT

The epithelia are the avascular layers of cells that cover the environment-exposed surfaces of the body. It appears that T cells localize to selected sites in or adjacent to epithelia via the selective expression of adhesion molecules and chemokine receptors on T cells. These bind to counter-receptors and to chemokines expressed by epithelial cells. Recently, there has been an advance in our understanding of the interaction of the alpha(Ebeta7) integrin with its epithelial cell ligand, E-cadherin. In addition, a new adhesion molecule has been identified on non-intestinal epithelial cells, termed lymphocyte-endothelial-epithelial-cell adhesion molecule (LEEP-CAM). Finally, there have been advances in our understanding of the role of skin- or gut-epithelia-derived chemokines in regulating activated T cell homing to these sites.


Subject(s)
Cell Communication , Epithelial Cells/metabolism , T-Lymphocytes/metabolism , Animals , Cadherins/metabolism , Cell Adhesion Molecules/metabolism , Chemokines/metabolism , Epithelial Cells/cytology , Humans , Integrins/metabolism , Membrane Glycoproteins/metabolism , Protein Binding , T-Lymphocytes/cytology
11.
J Exp Med ; 192(5): 761-8, 2000 Sep 04.
Article in English | MEDLINE | ID: mdl-10974041

ABSTRACT

The immune system has evolved specialized cellular and molecular mechanisms for targeting and regulating immune responses at epithelial surfaces. Here we show that small intestinal intraepithelial lymphocytes and lamina propria lymphocytes migrate to thymus-expressed chemokine (TECK). This attraction is mediated by CC chemokine receptor (CCR)9, a chemoattractant receptor expressed at high levels by essentially all CD4(+) and CD8(+) T lymphocytes in the small intestine. Only a small subset of lymphocytes in the colon are CCR9(+), and lymphocytes from other tissues including tonsils, lung, inflamed liver, normal or inflamed skin, inflamed synovium and synovial fluid, breast milk, and seminal fluid are universally CCR9(-). TECK expression is also restricted to the small intestine: immunohistochemistry reveals that intense anti-TECK reactivity characterizes crypt epithelium in the jejunum and ileum, but not in other epithelia of the digestive tract (including stomach and colon), skin, lung, or salivary gland. These results imply a restricted role for lymphocyte CCR9 and its ligand TECK in the small intestine, and provide the first evidence for distinctive mechanisms of lymphocyte recruitment that may permit functional specialization of immune responses in different segments of the gastrointestinal tract. Selective expression of chemokines by differentiated epithelium may represent an important mechanism for targeting and specialization of immune responses.


Subject(s)
Chemokines, CC/analysis , Intestinal Mucosa/immunology , Intestine, Small/immunology , Receptors, Chemokine/analysis , Animals , Chemokines, CC/physiology , Humans , Mice , Organ Specificity , Receptors, CCR , Receptors, Chemokine/physiology , T-Lymphocytes/chemistry
12.
Curr Biol ; 10(6): 325-8, 2000 Mar 23.
Article in English | MEDLINE | ID: mdl-10744978

ABSTRACT

HIV particles that use the chemokine receptor CXCR4 as a coreceptor for entry into cells (X4-HIV) inefficiently transmit infection across mucosal surfaces [1], despite their presence in seminal fluid and mucosal secretions from infected individuals [2] [3] [4]. In addition, although intestinal lymphocytes are susceptible to infection with either X4-HIV particles or particles that use the chemokine receptor CCR5 for viral entry (R5-HIV) during ex vivo culture [5], only systemic inoculation of R5-chimeric simian-HIV (S-HIV) results in a rapid loss of CD4(+) intestinal lymphocytes in macaques [6]. The mechanisms underlying the inefficient capacity of X4-HIV to transmit infection across mucosal surfaces and to infect intestinal lymphocytes in vivo have remained elusive. The CCR5 ligands RANTES, MIP-1alpha and MIP-1beta suppress infection by R5-HIV-1 particles via induction of CCR5 internalization, and individuals whose peripheral blood lymphocytes produce high levels of these chemokines are relatively resistant to infection [7] [8] [9]. Here, we show that the CXCR4 ligand stromal derived factor-1 (SDF-1) is constitutively expressed by mucosal epithelial cells at sites of HIV transmission and propagation. Furthermore, CXCR4 is selectively downmodulated on intestinal lymphocytes within the setting of prominent SDF-1 expression. We postulate that mucosally derived SDF-1 continuously downmodulates CXCR4 on resident HIV target cells, thereby reducing the transmission and propagation of X4-HIV at mucosal sites. Moreover, such a mechanism could contribute to the delayed emergence of X4 isolates, which predominantly occurs during the later stages of the HIV infection.


Subject(s)
Chemokines, CXC/physiology , HIV/growth & development , Intestinal Mucosa/metabolism , Chemokine CXCL12 , Chemokines, CXC/biosynthesis , Humans , Receptors, CCR5/metabolism , Receptors, CXCR4/biosynthesis , Receptors, CXCR4/genetics
13.
Eur J Immunol ; 30(3): 819-26, 2000 Mar.
Article in English | MEDLINE | ID: mdl-10741397

ABSTRACT

To determine which chemokine receptors might be involved in T lymphocyte localization to the intestinal mucosa, we examined receptor expression on human intestinal lamina propria lymphocytes (LPL), intraepithelial lymphocytes (IEL) and CD45RO+beta7hi gut homing peripheral blood lymphocytes (PBL). Virtually all LPL and IEL expressed CXCR3 and CCR5, receptors that have been associated with Th1(Tc1)/Th0 lymphocytes, while CCR3 and CCR4, receptors associated with Th2 (Tc2)lymphocytes, CCR7, CXCR1 and CXCR2 were not expressed. CXCR3 and CCR5 receptors were functional, as LPL and IEL migrated to their respective ligands I-TAC and RANTES. In addition, most alphaEbeta7- LPL and IEL expressed high levels of CCR2. While the majority of CD45RO(-)beta7hi PBL also expressed CXCR3 and CCR5, a proportion of these cells were CXCR3- and/or CCR5- and some expressed CCR4 and/or CCR7, indicating that lymphocytes recruited to the intestinal mucosa represent a subset of these cells. In summary, our results show that LPL and IEL within the normal intestine express a specific and similar array of chemokine receptors whose ligands are constitutively expressed in the intestinal mucosa and whose expression is up-regulated during intestinal inflammation. These results support the view that CXCR3, CCR5 and CCR2 may play an important role in lymphocyte localization within the intestinal mucosa.


Subject(s)
Integrin beta Chains , Intestinal Mucosa/cytology , Intestinal Mucosa/immunology , Lymphocytes/immunology , Receptors, Chemokine/metabolism , Animals , Antibodies, Monoclonal , Chemotaxis, Leukocyte , Epithelial Cells/cytology , Epithelial Cells/immunology , Humans , In Vitro Techniques , Inflammation/immunology , Inflammation/pathology , Integrins/metabolism , Leukocyte Common Antigens/metabolism , Mice , Receptors, CCR2 , Receptors, CCR5/metabolism , Receptors, CXCR3 , T-Lymphocytes/immunology
14.
J Exp Med ; 190(9): 1241-56, 1999 Nov 01.
Article in English | MEDLINE | ID: mdl-10544196

ABSTRACT

TECK (thymus-expressed chemokine), a recently described CC chemokine expressed in thymus and small intestine, was found to mediate chemotaxis of human G protein-coupled receptor GPR-9-6/L1.2 transfectants. This activity was blocked by anti-GPR-9-6 monoclonal antibody (mAb) 3C3. GPR-9-6 is expressed on a subset of memory alpha4beta7(high) intestinal trafficking CD4 and CD8 lymphocytes. In addition, all intestinal lamina propria and intraepithelial lymphocytes express GPR-9-6. In contrast, GPR-9-6 is not displayed on cutaneous lymphocyte antigen-positive (CLA(+)) memory CD4 and CD8 lymphocytes, which traffic to skin inflammatory sites, or on other systemic alpha4beta7(-)CLA(-) memory CD4/CD8 lymphocytes. The majority of thymocytes also express GPR-9-6, but natural killer cells, monocytes, eosinophils, basophils, and neutrophils are GPR-9-6 negative. Transcripts of GPR-9-6 and TECK are present in both small intestine and thymus. Importantly, the expression profile of GPR-9-6 correlates with migration to TECK of blood T lymphocytes and thymocytes. As migration of these cells is blocked by anti-GPR-9-6 mAb 3C3, we conclude that GPR-9-6 is the principal chemokine receptor for TECK. In agreement with the nomenclature rules for chemokine receptors, we propose the designation CCR-9 for GPR-9-6. The selective expression of TECK and GPR-9-6 in thymus and small intestine implies a dual role for GPR-9-6/CCR-9, both in T cell development and the mucosal immune response.


Subject(s)
Chemokines, CC/pharmacology , Chemotaxis/immunology , Intestinal Mucosa/immunology , Receptors, Chemokine/immunology , T-Lymphocytes/metabolism , Thymus Gland/immunology , Antibodies, Monoclonal , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/metabolism , Calcium , Cell Line , Chemokines, CC/genetics , Flow Cytometry , Gene Expression Regulation/immunology , Humans , Lymphocyte Activation/immunology , Lymphocyte Subsets/immunology , RNA, Messenger/immunology , Receptors, CCR , Receptors, Chemokine/genetics , Receptors, Lymphocyte Homing/immunology , Transfection
15.
J Immunol ; 163(3): 1592-601, 1999 Aug 01.
Article in English | MEDLINE | ID: mdl-10415064

ABSTRACT

Upon encountering the relevant vascular bed, lymphocytes attach to endothelial adhesion molecules, transmigrate out of circulation, and localize within tissues. Lymphocytes may then be retained at microanatomic sites, as in tissues, or they may continue to migrate to the lymphatics and recirculate in the blood. Lymphocytes also interact transiently, but with high avidity, with target cells or APC that are infected with microbes or have taken up exogenous foreign Ags. This array of adhesive capabilities is mediated by the selective expression of lymphocyte adhesion molecules. Here, we developed the 6F10 mAb, which recognizes a cell surface glycoprotein designated lymphocyte endothelial-epithelial cell adhesion molecule (LEEP-CAM), that is distinct in biochemical characteristics and distribution of expression from other molecules known to play a role in lymphocyte adhesion. LEEP-CAM is expressed on particular epithelia, including the suprabasal region of the epidermis, the basal layer of bronchial and breast epithelia, and throughout the tonsillar and vaginal epithelia. Yet, it is absent from intestinal and renal epithelia. Interestingly, it is expressed also on vascular endothelium, especially high endothelial venules (HEV) in lymphoid organs, such as tonsil and appendix. The anti-LEEP-CAM mAb specifically blocked T and B lymphocyte adhesion to monolayers of epithelial cells and to vascular endothelial cells in static cell-to-cell binding assays by approximately 40-60% when compared with control mAbs. These data suggest a role for this newly identified molecule in lymphocyte binding to endothelium, as well as adhesive interactions within selected epithelia.


Subject(s)
Cell Adhesion Molecules/physiology , Endothelium, Vascular/physiology , Epithelial Cells/immunology , Lymphocytes/physiology , Membrane Glycoproteins/physiology , Antibodies, Blocking/chemistry , Antibodies, Blocking/metabolism , Antibodies, Blocking/physiology , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/metabolism , Antibodies, Monoclonal/physiology , Binding Sites, Antibody , Binding, Competitive/immunology , Cations, Divalent/chemistry , Cell Adhesion/immunology , Cell Adhesion Molecules/chemistry , Cell Adhesion Molecules/immunology , Cell Line , Endothelium, Vascular/chemistry , Endothelium, Vascular/immunology , Glycoside Hydrolases/metabolism , Humans , Intestinal Mucosa , Leukocytes/metabolism , Lymphocytes/immunology , Membrane Glycoproteins/chemistry , Membrane Glycoproteins/immunology , Organ Specificity/immunology , Precipitin Tests , Staining and Labeling
16.
J Immunol ; 162(11): 6641-9, 1999 Jun 01.
Article in English | MEDLINE | ID: mdl-10352281

ABSTRACT

The mucosal lymphocyte integrin alpha E(CD103)beta 7 is thought to be important for intraepithelial lymphocyte (IEL) localization or function. We cloned the murine integrin gene encoding alpha E, localized it to chromosome 11, and generated integrin alpha E-deficient mice. In alpha E-/- mice, intestinal and vaginal IEL numbers were reduced, consistent with the known binding of alpha E beta 7 to E-cadherin expressed on epithelial cells. However, it was surprising that lamina propria T lymphocyte numbers were diminished, as E-cadherin is not expressed in the lamina propria. In contrast, peribronchial, intrapulmonary, Peyer's patch, and splenic T lymphocyte numbers were not reduced in alpha E-deficient mice. Thus, alpha E beta 7 was important for generating or maintaining the gut and vaginal T lymphocytes located diffusely within the epithelium or lamina propria but not for generating the gut-associated organized lymphoid tissues. Finally, the impact of alpha E deficiency upon intestinal IEL numbers was greater at 3-4 wk of life than in younger animals, and affected the TCR alpha beta+ CD8+ T cells more than the gamma delta T cells or the TCR alpha beta+ CD4+CD8- population. These findings suggest that alpha E beta 7 is involved in the expansion/recruitment of TCR alpha beta+ CD8+ IEL following microbial colonization. Integrin alpha E-deficient mice will provide an important tool for studying the role of alpha E beta 7 and of alpha E beta 7-expressing mucosal T lymphocytes in vivo.


Subject(s)
Antigens, CD/genetics , Integrin alpha Chains , Intestinal Mucosa/immunology , Intestinal Mucosa/pathology , Lymphopenia/genetics , Lymphopenia/immunology , T-Lymphocyte Subsets/immunology , Animals , CD4-Positive T-Lymphocytes/pathology , CD8-Positive T-Lymphocytes/pathology , Chromosome Mapping , Cloning, Molecular , Crosses, Genetic , Epithelial Cells/immunology , Epithelial Cells/pathology , Female , Hyaluronan Receptors/biosynthesis , Intestinal Mucosa/metabolism , Lymphocyte Count , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Molecular Sequence Data , Muridae , Peyer's Patches/cytology , Peyer's Patches/immunology , Receptors, Antigen, T-Cell, alpha-beta/metabolism , Spleen/cytology , Spleen/pathology , T-Lymphocyte Subsets/metabolism , T-Lymphocyte Subsets/pathology , Vagina/immunology , Vagina/pathology
17.
J Exp Med ; 189(10): 1631-8, 1999 May 17.
Article in English | MEDLINE | ID: mdl-10330442

ABSTRACT

The requirement of beta7 integrins for lymphocyte migration was examined during an ongoing immune response in vivo. Transgenic mice (OT-I) expressing an ovalbumin-specific major histocompatibility complex class I-restricted T cell receptor for antigen were rendered deficient in expression of all beta7 integrins or only the alphaEbeta7 integrin. To quantitate the relative use of beta7 integrins in migration in vivo, equal numbers of OT-I and OT-I-beta7(-/-) or OT-I-alphaE-/- lymph node (LN) cells were adoptively transferred to normal mice. Although OT-I-beta7(-/-) LN cells migrated to mesenteric LN and peripheral LN as well as wild-type cells, beta7 integrins were required for naive CD8 T cell and B cell migration to Peyer's patch. After infection with a recombinant virus (vesicular stomatitis virus) encoding ovalbumin, beta7 integrins became critical for migration of activated CD8 T cells to the mesenteric LN and Peyer's patch. Naive CD8 T cells did not enter the lamina propria or the intestinal epithelium, and the majority of migration of activated CD8 T cells to the small and large intestinal mucosa, including the epithelium, was beta7 integrin-mediated. The alphaEbeta7 integrin appeared to play no role in migration during a primary CD8 T cell immune response in vivo. Furthermore, despite dramatic upregulation of alphaEbeta7 by CD8 T cells after entry into the epithelium, long-term retention of intestinal intraepithelial lymphocytes was also alphaEbeta7 independent.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Integrin beta Chains , Integrins/immunology , Lymph Nodes/immunology , Animals , CD8-Positive T-Lymphocytes/virology , Cell Movement , Mice , Mice, Transgenic , Ovalbumin , Peyer's Patches/immunology , Receptors, Antigen, T-Cell/immunology , Up-Regulation/immunology , Vesicular stomatitis Indiana virus/immunology
18.
J Cell Biol ; 140(1): 197-210, 1998 Jan 12.
Article in English | MEDLINE | ID: mdl-9425167

ABSTRACT

The cadherins are a family of homophilic adhesion molecules that play a vital role in the formation of cellular junctions and in tissue morphogenesis. Members of the integrin family are also involved in cell to cell adhesion, but bind heterophilically to immunoglobulin superfamily molecules such as intracellular adhesion molecule (ICAM)-1, vascular cell adhesion molecule (VCAM)-1, or mucosal addressin cell adhesion molecule (MadCAM)-1. Recently, an interaction between epithelial (E-) cadherin and the mucosal lymphocyte integrin, alphaEbeta7, has been proposed. Here, we demonstrate that a human E-cadherin-Fc fusion protein binds directly to soluble recombinant alphaEbeta7, and to alphaEbeta7 solubilized from intraepithelial T lymphocytes. Furthermore, intraepithelial lymphocytes or transfected JY' cells expressing the alphaEbeta7 integrin adhere strongly to purified E-cadherin-Fc coated on plastic, and the adhesion can be inhibited by antibodies to alphaEbeta7 or E-cadherin. The binding of alphaEbeta7 integrin to cadherins is selective since cell adhesion to P-cadherin-Fc through alphaEbeta7 requires >100-fold more fusion protein than to E-cadherin-Fc. Although the structure of the alphaE-chain is unique among integrins, the avidity of alphaEbeta7 for E-cadherin can be regulated by divalent cations or phorbol myristate acetate. Cross-linking of the T cell receptor complex on intraepithelial lymphocytes increases the avidity of alphaEbeta7 for E-cadherin, and may provide a mechanism for the adherence and activation of lymphocytes within the epithelium in the presence of specific foreign antigen. Thus, despite its dissimilarity to known integrin ligands, the specific molecular interaction demonstrated here indicates that E-cadherin is a direct counter receptor for the alphaEbeta7 integrin.


Subject(s)
Cadherins/physiology , Integrins/physiology , Amino Acid Sequence , Animals , Antibodies, Monoclonal , Cadherins/biosynthesis , Cadherins/chemistry , Cell Adhesion , Cell Line , Genetic Vectors , Humans , Immunoglobulin Fc Fragments/chemistry , Immunoglobulin G/chemistry , Integrins/biosynthesis , Intestinal Mucosa , Kinetics , Mice , Models, Molecular , Polymerase Chain Reaction , Protein Binding , Recombinant Fusion Proteins/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/physiology , Transfection
20.
AORN J ; 66(3): 405, 408-11, 413-4, 1997 Sep.
Article in English | MEDLINE | ID: mdl-9293322

ABSTRACT

The use of powered instrumentation in functional endoscopic sinus surgery has been a revolutionary development in the surgical treatment of chronic sinusitis. Several studies have demonstrated the safety, efficacy, and ease of use of this new technique. To provide support and coordinate the surgical process in powered functional endoscopic sinus surgery procedures, perioperative nurses must have an appreciation for its specific equipment handling and for appropriate patient care. This article describes a specific protocol that perioperative nurses can use to facilitate efficient and safe surgical environments for patients who undergo powered endoscopic sinus surgery procedures.


Subject(s)
Endoscopy/methods , Endoscopy/nursing , Perioperative Nursing , Sinusitis/surgery , Chronic Disease , Electric Power Supplies , Endoscopy/adverse effects , Humans , United States
SELECTION OF CITATIONS
SEARCH DETAIL