Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
1.
Isr Med Assoc J ; 19(10): 635-639, 2017 Oct.
Article in English | MEDLINE | ID: mdl-29103242

ABSTRACT

BACKGROUND: Invasive cervical cancer is caused by human papillomavirus (HPV). OBJECTIVES: To describe the prevalence and genotype distribution of HPV types in women at risk for cervical neoplasia. METHODS: Our study summarized HPV types detected in 6654 samples that were sent to the serology laboratory from cervical clinics in northern Israel between 2006-2014. The HPV test was performed during investigation of atypical squamous cells of undetermined significance (ASCUS) results on Pap tests or due to complaints suggestive of cervical neoplasia. HPV types were classified as high risk (HPV-HR) and low risk (HPV-LR). RESULTS: Of the samples, 46.4% (3085/6654) were HPV-HR positive. Of women with cervical intraepithelial neoplasia 2-3 (CIN 2-3) or cancer, 292/318 (91.8%) and 137/145 (94.5%), respectively, were HPV-HR positive. HPV 16 and HPV 18 were detected in 11.8% of the total samples and in 48.2% and 64.9% of the women with CIN 2-3 and with cancer, respectively. HPV was negative in 8/145 (5.5%) and 26/318 (8.2%) of women with cervical cancer and CIN 2-3, respectively. CONCLUSIONS: This study shows the prevalence of HPV types in women at risk for cervical neoplasia. The sensitivity of all HPV types for CIN 2-3 and cervical cancer was 91.8% and 94.5%, respectively; and of HPV-HR types, 89% and 92.4%, respectively. Triage of HPV-HR types should be considered in women with ASCUS because HPV-HR types were discovered in only 36.7%. The distribution of HPV types in our population is similar to that reported for other developed countries.


Subject(s)
DNA, Viral/analysis , Papillomaviridae , Papillomavirus Infections , Uterine Cervical Dysplasia , Uterine Cervical Neoplasms , Adult , Female , Genotype , Humans , Israel/epidemiology , Middle Aged , Papanicolaou Test , Papillomaviridae/genetics , Papillomaviridae/isolation & purification , Papillomavirus Infections/complications , Papillomavirus Infections/diagnosis , Papillomavirus Infections/epidemiology , Prevalence , Retrospective Studies , Risk Assessment/methods , Serologic Tests/methods , Serologic Tests/statistics & numerical data , Uterine Cervical Neoplasms/epidemiology , Uterine Cervical Neoplasms/pathology , Uterine Cervical Neoplasms/virology , Uterine Cervical Dysplasia/epidemiology , Uterine Cervical Dysplasia/pathology , Uterine Cervical Dysplasia/virology
2.
Oncoimmunology ; 5(2): e1078056, 2016 Feb.
Article in English | MEDLINE | ID: mdl-27057452

ABSTRACT

Extracellular matrix metalloproteinase inducer (EMMPRIN/CD147) mediates tumor cell-macrophage interactions, and has been shown to induce both matrix metalloproteinases (MMPs) and vascular endothelial growth factor (VEGF). However, the epitope responsible for MMP induction is controversial, and the epitope responsible for VEGF induction is yet unknown. We generated a novel anti-EMMPRIN antibody directed against a specific epitope that successfully inhibited the production of both MMP-9 and VEGF in tumor cell-macrophage in vitro co-culture systems, exhibiting a U-shaped dose response. Furthermore, this antibody efficiently inhibited in vivo tumor progression in both the RENCA renal cell carcinoma and CT26 colon carcinoma subcutaneous tumor models, and reduced tumor size and number of metastatic foci in the 4T1 orthotopic model. This was achieved by inhibiting angiogenesis as assessed by immunohistochemical staining for the endothelial marker CD31, by inhibiting tumor cell proliferation as assessed by the staining for Ki-67, and by enhancing tumor cell apoptosis as assessed in the TUNEL assay. Moreover, administration of the antibody recruited more macrophages into the tumor, and skewed the tumor microenvironment for macrophages from TGFß-dominated anti-inflammatory microenvironment, to a less immunosuppressive one. The antibody improved the ability of stimulated macrophages to perform antibody-dependent cell cytotoxicity (ADCC) and kill tumor cells. Thus, our new antibody maps the epitope capable of inducing both MMPs and VEGF, and places EMMPRIN as a good target for cancer therapy.

3.
Shock ; 37(1): 95-102, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21921827

ABSTRACT

We evaluated the effects of hyperoxia on pulmonary inflammatory changes in sepsis induced by cecal ligation and puncture (CLP) in rats. Seven groups were studied: sham-operated rats breathing air for 20 or 48 h; CLP breathing air for 20 or 48 h; and CLP + 100% oxygen for 20 h, or 70% oxygen for 48 h, or 100% oxygen intermittently (6 h/d) for 48 h. Video microscopy was used to monitor lung macromolecular leak, microvascular flow velocity, and shear rates, and lung morphometry was used for leukocyte infiltration and solid tissue area. Cell counts, tumor necrosis factor α, and nitrites were determined in peripheral blood and lung lavage fluid. Expression of adhesion molecules in blood leukocytes was evaluated by flow cytometry. Cecal ligation and puncture induced inflammation manifested in leukopenia, left shift, thrombocytopenia, increased expression of L selectin and CD11, increased serum and lavage fluid tumor necrosis factor α and leukocytes, and increased lung tissue area, macromolecular leak, and sequestration of leukocytes. Inhalation of 100% oxygen for 20 h increased nitrites (P < 0.01) and decreased leukocyte count in lavage fluid (P < 0.05) and attenuated lung macromolecular leak and changes in solid tissue area (P < 0.01). Inhalation of 70% oxygen (48 h) attenuated expression of adhesion molecules (P < 0.001) but failed to attenuate markers of lung inflammation. In contrast, intermittent 100% oxygen exerted favorable effects on markers of inflammation, attenuated leukocyte expression of L selectin and CD11 (P < 0.01), decreased pulmonary sequestration of leukocytes (P < 0.001), and ameliorated changes in macromolecular leak (P < 0.01) and lung solid tissue area (P < 0.05). Our data support the beneficial effects of safe subtoxic regimens of normobaric hyperoxia on the systemic and pulmonary inflammatory response following CLP.


Subject(s)
Hyperoxia/metabolism , Lung/metabolism , Oxygen/pharmacology , Pneumonia/metabolism , Sepsis/metabolism , Animals , CD11 Antigens/biosynthesis , CD11 Antigens/immunology , Disease Models, Animal , Dose-Response Relationship, Drug , Gene Expression Regulation/drug effects , Hyperoxia/complications , Hyperoxia/immunology , Hyperoxia/pathology , L-Selectin/biosynthesis , L-Selectin/immunology , Leukocytes/immunology , Leukocytes/metabolism , Leukocytes/pathology , Lung/immunology , Lung/pathology , Male , Nitrites/immunology , Nitrites/metabolism , Oxygen/metabolism , Pneumonia/complications , Pneumonia/immunology , Rats , Rats, Sprague-Dawley , Sepsis/complications , Sepsis/immunology , Tumor Necrosis Factor-alpha/metabolism
4.
Transpl Int ; 24(10): 1018-26, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21806687

ABSTRACT

Transplantation involves preoperative ischemic periods that contribute to endothelial cell (EC) dysfunction and T-cell activation, leading to graft rejection. As hypoxia is a major constituent of ischemia, we evaluated its effect on the ability of ECs to express HLA-DR, which is required for presentation of antigens to T cells, and by itself serves as an important target for allogeneic T cells. Primary human umbilical vein ECs (HUVEC) and the human endothelial cell line EaHy926 were incubated in normoxia or hypoxia (PO(2) < 0.3%). Hypoxia increased the membranal expression (by 4-6 fold, P < 0.01) and secretion (by sixfold, P < 0.05) of HLA-DR protein, without influencing the accumulation of its mRNA. Alternative splicing, attenuated trafficking, or shedding from the plasma membrane were not observed, but the lysosomal inhibitor bafilomycin A1 reduced HLA-DR secretion. Hypoxia-induced endothelial HLA-DR elevated and diminished the secretion of IL-2 and IL-10, respectively, from co-cultured allogeneic CD4(+) T cells in a HLA-DR-dependent manner, as demonstrated by the use of monoclonal anti-HLA-DR. Our results indicate a yet not fully understood post-translational mechanism(s), which elevate both membranal and soluble HLA-DR expression. This elevation is involved in allogeneic T-cell activation, highlighting the pivotal role of ECs in ischemia/hypoxia-associated injury and graft rejection.


Subject(s)
Endothelial Cells/cytology , HLA-DR Antigens/biosynthesis , Hypoxia , Lymphocyte Activation , T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/immunology , Cell Line , Graft Rejection , HLA-DR Antigens/metabolism , Humans , Hypoxia/metabolism , Inflammation , Oxygen/metabolism , Radioimmunoassay/methods
5.
Graefes Arch Clin Exp Ophthalmol ; 249(7): 1047-55, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21452038

ABSTRACT

OBJECTIVE: Glaucoma filtering surgery may be compromised by cystic blebs which develop more frequently when anti-metabolites are used to arrest wound healing. Matrix metalloproteinases (MMPs) and the naturally occurring tissue inhibitors of metalloproteinases (TIMPs) are essential in connective tissue remodeling and wound healing. This study aimed to determine whether filtering blebs display increased expression of MMP-2, MMP-9, TIMP-1 and TIMP-2, and whether it is reflected in tear fluid. METHODS: Tissue samples from leaking blebs (n = 5) and control conjunctiva (n = 5) were evaluated by immunohistochemistry for MMP-2, MMP-9, TIMP-1 and TIMP-2. Tear fluid was collected from 12 patients (12 eyes) with cystic blebs and ten patients (ten eyes) with flat blebs following trabeculectomy with Mitomycin C applied and 16 controls. MMP levels were evaluated by zymography and TIMP levels by Western blot analysis. RESULTS: Conjunctival tissue was obtained from five eyes with cystic leaking blebs and five control eyes undergoing cataract surgery. More extensive MMP-2 and MMP-9 expression was found in the epithelial and stromal layers of blebs than in control conjunctiva. TIMP-1and TIMP-2 were expressed in all layers of the blebs, but only in the epithelium of control conjunctiva. MMP-2 and proMMP-2 activity in tears from eyes with flat blebs was significantly higher than that of controls, while activity in tears of eyes with cystic blebs was significantly higher than in those with flat blebs. There was no difference in MMP-9 activity between tears of control and post-filtering surgery eyes. CONCLUSIONS: Increased MMPs and TIMPs expression is associated with the formation of filtering blebs, suggesting involvement of MMPs in bleb remodeling. MMP-2 and ProMMP-2 levels in tear fluid may be markers for bleb configuration.


Subject(s)
Conjunctiva/physiology , Eye Proteins/metabolism , Glaucoma, Open-Angle/enzymology , Metalloproteases/metabolism , Tears/enzymology , Trabeculectomy , Aged , Aged, 80 and over , Alkylating Agents/administration & dosage , Blotting, Western , Conjunctiva/surgery , Female , Fistula/enzymology , Glaucoma, Open-Angle/surgery , Humans , Immunoenzyme Techniques , Male , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism , Middle Aged , Mitomycin/administration & dosage , Tissue Inhibitor of Metalloproteinase-1/metabolism , Tissue Inhibitor of Metalloproteinase-2/metabolism , Wound Healing/drug effects
6.
Am J Physiol Cell Physiol ; 300(3): C557-66, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21148412

ABSTRACT

Hypoxia, which characterizes ischemia, trauma, inflammation, and solid tumors, recruits monocytes, immobilizes them, and alters their function, leading to an anti-inflammatory and proangiogenic phenotype. Monocyte extravasation from the circulation and their migration in tissues are partially mediated by the balance between matrix metalloproteinases (MMPs) and their tissue inhibitors (TIMPs). The mechanisms evoked by hypoxia that regulate monocyte migration and activation are not entirely clear. Specifically, the effect of hypoxia on TIMPs in these cells has hardly been investigated. We show that hypoxia reduces TIMP-2 secretion from human primary monocytes and from the monocyte-like cell lines U937 and THP-1 by three- to fourfold (P < 0.01), by inhibiting TIMP-2 transcription through mechanisms that involve the transcription factor SP-1. Hypoxia also lowers TIMP-2 protein secretion from human endothelial cells (by 2-fold, P < 0.05). TIMP-2 levels do not influence the reduced migration of THP-1 cells in hypoxia; however, low TIMP-2 levels enhance endothelial cell migration/proliferation, their ability to form tubelike structures in vitro, and the appearance of mature blood vessels in a Matrigel plug assay in vivo. Thus we conclude that reduced TIMP-2 levels secreted from both hypoxic monocytes and endothelial cells are proangiogenic.


Subject(s)
Down-Regulation , Hypoxia/metabolism , Monocytes/metabolism , Neovascularization, Physiologic , Tissue Inhibitor of Metalloproteinase-2/antagonists & inhibitors , Up-Regulation , Animals , Base Sequence , Cell Line , Cell Migration Inhibition/genetics , Cell Movement/genetics , Cell Proliferation , Cells, Cultured , Down-Regulation/genetics , Endothelial Cells/metabolism , Endothelial Cells/pathology , Humans , Hypoxia/pathology , Hypoxia/physiopathology , Mice , Mice, Inbred BALB C , Molecular Sequence Data , Monocytes/pathology , Neovascularization, Physiologic/genetics , Neovascularization, Physiologic/physiology , Sp1 Transcription Factor/physiology , Tissue Inhibitor of Metalloproteinase-2/biosynthesis , Tissue Inhibitor of Metalloproteinase-2/genetics , U937 Cells , Up-Regulation/genetics
7.
Front Immunol ; 2: 45, 2011.
Article in English | MEDLINE | ID: mdl-22566835

ABSTRACT

Monocytes and Macrophages (Mo/Mɸ) exhibit great plasticity, as they can shift between different modes of activation and, driven by their immediate microenvironment, perform divergent functions. These include, among others, patrolling their surroundings and maintaining homeostasis (resident Mo/Mɸ), combating invading pathogens and tumor cells (classically activated or M1 Mo/Mɸ), orchestrating wound healing (alternatively activated or M2 Mo/Mɸ), and restoring homeostasis after an inflammatory response (resolution Mɸ). Hypoxia is an important factor in the Mɸ microenvironment, is prevalent in many physiological and pathological conditions, and is interdependent with the inflammatory response. Although Mo/Mɸ have been studied in hypoxia, the mechanisms by which hypoxia influences the different modes of their activation, and how it regulates the shift between them, remain unclear. Here we review the current knowledge about the molecular mechanisms that mediate this hypoxic regulation of Mɸ activation. Much is known about the hypoxic transcriptional regulatory network, which includes the master regulators hypoxia-induced factor-1 and NF-κB, as well as other transcription factors (e.g., AP-1, Erg-1), but we also highlight the role of post-transcriptional and post-translational mechanisms. These mechanisms mediate hypoxic induction of Mɸ pro-angiogenic mediators, suppress M1 Mɸ by post-transcriptionally inhibiting pro-inflammatory mediators, and help shift the classically activated Mɸ into an activation state which approximate the alternatively activated or resolution Mɸ.

8.
Am J Pathol ; 177(4): 2046-54, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20709800

ABSTRACT

Tumor-associated macrophages can potentially kill tumor cells via the high concentrations of nitric oxide (NO) produced by inducible nitric oxide synthase (iNOS); however, tumor-associated macrophages actually support tumor growth, as they are skewed toward M2 activation, which is characterized by low amounts of NO production and is proangiogenic. We show that the mouse renal cell carcinoma cell line, RENCA, which, on stimulation, expresses high levels of iNOS mRNA, loses its ability to express the iNOS protein. This effect is mediated by the microRNA miR-146a, as inhibition of RENCA cells with anti-miR- 146a restores iNOS expression and NO production (4.8 ± 0.4 versus 0.3 ± 0.1 µmol/L in uninhibited cells, P < 0.001). In vivo, RENCA tumor cells do not stain for iNOS, while infiltrating tumor-associated macrophages showed intense staining, and both cell types expressed iNOS mRNA. Restoring iNOS protein expression in RENCA cells using anti-miR-146a increases macrophage-induced death of RENCA cells by 73% (P < 0.01) in vitro and prevents tumor growth in vivo. These results suggest that, in addition to NO production by macrophages, tumor cells must produce NO to induce their own deaths, and some tumor cells may use miR-146a to reduce or abolish endogenous NO production to escape macrophage-mediated cell death. Thus, inhibiting miR-146a may render these tumor cells susceptible to therapeutic strategies, such as adoptive transfer of M1-activated macrophages.


Subject(s)
Carcinoma, Renal Cell/pathology , Kidney Neoplasms/pathology , MicroRNAs/pharmacology , Nitric Oxide Synthase Type II/antagonists & inhibitors , Nitric Oxide Synthase Type II/metabolism , Animals , Apoptosis , Blotting, Western , Carcinoma, Renal Cell/enzymology , Carcinoma, Renal Cell/genetics , Cell Movement , Cell Proliferation , Female , In Situ Hybridization , Kidney Neoplasms/enzymology , Kidney Neoplasms/genetics , Macrophages/enzymology , Macrophages/pathology , Mice , Mice, Inbred BALB C , MicroRNAs/antagonists & inhibitors , Neovascularization, Pathologic , Nitric Oxide/metabolism , Nitric Oxide Synthase Type II/genetics , Protein Biosynthesis , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction
9.
Isr Med Assoc J ; 12(3): 158-62, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20684180

ABSTRACT

BACKGROUND: Interleukin-10 is an anti-inflammatory cytokine and consequently is considered by many to have a protective role in heart failure, as opposed to the notorious tumor necrosis factor-alpha. OBJECTIVES: To test the hypothesis of the possible beneficial impact of IL-10 on mortality in systolic heart failure patients in relation to their circulating TNFalpha levels. METHODS: We measured circulating levels of IL-10 and TNFalpha in 67 ambulatory systolic heart failure patients (age 65 +/- 13 years). RESULTS: Mortality was or tended to be higher in patients with higher levels (above median level) of circulating TNFalpha (9/23, 39% vs. 6/44, 14%; P = 0.02) or IL-10 (10/34, 30% vs. 5/33, 15%; P = 0.10). However, mortality was highest in the subset of patients with elevation of both markers above median (7/16, 44% vs. 8/51, 16%; P = 0.019). Elevation of both markers was associated with more than a threefold hazard ratio for mortality (HR 3.67, 95% confidence interval 1.14-11.78). CONCLUSIONS: Elevated circulating IL-10 levels in systolic heart failure patients do not have a protective counterbalance effect on mortality. Moreover, patients with elevated IL-10 and TNFalpha had significantly higher mortality, suggesting that the possible interaction in the complex inflammatory and anti-inflammatory network may need further study.


Subject(s)
Heart Failure, Systolic/blood , Interleukin-10/blood , Tumor Necrosis Factor-alpha/blood , Aged , Body Mass Index , C-Reactive Protein/analysis , Cause of Death , Cholesterol/blood , Chronic Disease , Creatinine/blood , Echocardiography , Electrocardiography , Exercise Test , Female , Follow-Up Studies , Heart Failure, Systolic/diagnostic imaging , Hemoglobins/analysis , Humans , Male , Matrix Metalloproteinase 9/blood , Middle Aged , Natriuretic Peptide, Brain/blood , Peptide Fragments/blood , Protein Precursors/blood , Stroke Volume/physiology , Troponin T/blood , Ventricular Dysfunction, Left/blood , Ventricular Dysfunction, Left/diagnostic imaging
10.
Am J Physiol Cell Physiol ; 298(4): C942-51, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20071690

ABSTRACT

Detachment of endothelial cells (ECs) from the extracellular matrix (ECM) is required not only for angiogenesis, but also for EC apoptosis. Matrix metalloproteinase (MMP)-2 plays a major role in the degradation of the ECM, supporting an essential role for this enzyme in both survival (angiogenesis) and death of ECs. Our aim was to study these seemingly paradoxical effects of MMP-2. We rationalized that inhibiting apoptosis would drive MMP-2 toward a prosurvival activity, clarifying the mechanisms involved. By employing specific inhibitors to two major apoptotic pathways in ECs, caspases and p38 MAPK (p38), we demonstrated that they differently affected EC behavior as well as MMP-2 expression. The p38 pathway appears to enhance MMP-2 synthesis, its partial ("intermediate") and its full activation, probably via membrane type (MT)1-MMP, while caspases enhance MMP-2 synthesis and full activation but reduce MT1-MMP and MMP-2 intermediate form. Evaluation of the reciprocal influences of MMP-2 on ECs showed that the intermediate form supported survival and migration, and the fully active form led to cell death. In addition, a pro- and intermediate form-rich environment, even in the presence of the fully active form, exerted protective effects. Thus the seemingly conflicting effects of MMP-2 on EC survival may be explained by the ratio between the MMP-2 activation forms. A regulatory loop between active MMP-2 and p38 but not between MMP-2 and caspases was also observed, suggesting that MMP-2 is downstream to caspases where it serves as an "exterminator" molecule. Altogether, modification of caspase and p38 pathways, via changes of local MMP-2, affect survival and angiogenic steps in ECs.


Subject(s)
Apoptosis/physiology , Endothelial Cells/physiology , Matrix Metalloproteinase 2/metabolism , Amino Acid Chloromethyl Ketones/pharmacology , Apoptosis/drug effects , Caspase 3/genetics , Caspase 3/metabolism , Caspase Inhibitors , Cell Movement/physiology , Cell Shape , Cells, Cultured , Endothelial Cells/cytology , Endothelial Cells/drug effects , Enzyme Inhibitors/pharmacology , Extracellular Matrix/metabolism , Humans , Imidazoles/pharmacology , Matrix Metalloproteinase 2/genetics , Pyridines/pharmacology , Signal Transduction/drug effects , Signal Transduction/physiology , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , p38 Mitogen-Activated Protein Kinases/genetics , p38 Mitogen-Activated Protein Kinases/metabolism
11.
J Immunol ; 183(7): 4705-14, 2009 Oct 01.
Article in English | MEDLINE | ID: mdl-19752225

ABSTRACT

Inflammation and angiogenesis are pivotal processes in the progression of many diseases, including malignancies. A hypoxic microenvironment often results in a milieu of proinflammatory and proangiogenic cytokines produced by infiltrating cells. We assessed the role of macrophage-derived hypoxia-associated cytokines in promoting inflammation and angiogenesis. Supernatants of macrophages, stimulated under hypoxia with or without an inflammatory stimulus (LPS), promoted angiogenesis when incorporated into Matrigel plugs. However, neutralization of IL-1 in the supernatants, particularly IL-1beta, completely abrogated cell infiltration and angiogenesis in Matrigel plugs and reduced vascular endothelial growth factor (VEGF) levels by 85%. Similarly, supernatants from macrophages of IL-1beta knockout mice did not induce inflammatory or angiogenic responses. The importance of IL-1 signaling in the host was demonstrated by the dramatic reduction of inflammatory and angiogenic responses in Matrigel plugs that contained macrophage supernatants from control mice which had been implanted in IL-1 receptor type I knockout mice. Myeloid cells infiltrating into Matrigel plugs were of bone marrow origin and represented the major source of IL-1 and other cytokines/chemokines in the plugs. Cells of endothelial lineage were the main source of VEGF and were recruited mainly from neighboring tissues, rather than from the bone marrow. Using the aortic ring sprouting assay, it was shown that in this experimental system, IL-1 does not directly activate endothelial cell migration, proliferation and organization into blood vessel-like structures, but rather activates infiltrating cells to produce endothelial cell activating factors, such as VEGF. Thus, targeting IL-1beta has the potential to inhibit angiogenesis in pathological situations and may be of considerable clinical value.


Subject(s)
Angiogenic Proteins/physiology , Cell Migration Inhibition/immunology , Interleukin-1alpha/physiology , Interleukin-1beta/physiology , Macrophages, Peritoneal/immunology , Macrophages, Peritoneal/metabolism , Neovascularization, Physiologic/immunology , Angiogenic Proteins/antagonists & inhibitors , Angiogenic Proteins/deficiency , Animals , Cells, Cultured , Collagen/physiology , Drug Combinations , Endothelial Cells/cytology , Endothelial Cells/immunology , Endothelial Cells/metabolism , Inflammation Mediators/physiology , Interleukin-1alpha/deficiency , Interleukin-1alpha/genetics , Interleukin-1beta/antagonists & inhibitors , Interleukin-1beta/deficiency , Laminin/physiology , Lipopolysaccharides/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Myeloid Cells/cytology , Myeloid Cells/immunology , Myeloid Cells/metabolism , Neovascularization, Pathologic/immunology , Neovascularization, Pathologic/pathology , Neovascularization, Physiologic/genetics , Proteoglycans/physiology , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Vascular Endothelial Growth Factor A/biosynthesis
12.
Crit Care Med ; 37(3): 1054-61, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19237917

ABSTRACT

OBJECTIVE: Inhalation of oxygen improves the hemodynamic status and attenuates the inflammatory response after intestinal ischemia-reperfusion (IR). Yet, the use of hyperoxia is hindered by concerns that it could exacerbate reperfusion injury by increasing free radical formation. We examined the effect of hyperoxia on enterocyte turnover and intestinal preservation and rehabilitation following IR injury in rats. DESIGN: Animal study. SETTING: Research laboratory. SUBJECTS: Male Sprague-Dawley rats. INTERVENTIONS: Animals were assigned to four experimental groups: 1) Sham rats underwent laparotomy without vascular occlusion and breathed air, 2) Sham-oxygen rats underwent laparotomy without vascular occlusion and breathed 100% oxygen, 3) IR rats underwent occlusion of the superior mesenteric artery and portal vein for 30 minutes and breathed air, and 4) IR group treated with oxygen (IR-O2)rats underwent IR and breathed 100% oxygen starting 10 minutes before and continued for the first 6 hours after reperfusion. Intestinal structural changes, enterocyte proliferation, and enterocyte apoptosis were determined 24 hours after IR. MEASUREMENTS AND MAIN RESULTS: In IR rats, breathing 100% oxygen resulted in a significant decrease in Park's injury score in the ileum (p < 0.05 from untreated IR rats). Rats treated with oxygen also demonstrated a significant increase in mucosal weight (p < 0.05) and mucosal DNA (p < 0.05) in the jejunum and ileum, and an increase in villus height (p < 0.01), and crypt depth (p < 0.05) in the ileum. Enterocyte proliferation (assessed by bromodeoxyuridine uptake) was significantly decreased in the jejunum and ileum in untreated IR rats. Oxygen therapy increased enterocyte proliferation in the ileum, and diminished both the apoptosis index and Bax gene expression in the jejunum and ileum (p < 0.05). Plasma thermochemiluminescence oxidizability assay revealed significantly higher thermochemiluminescence ratios in IR group treated with oxygen than in untreated IR rats (p < 0.05) at 6 hours postreperfusion suggesting a significantly lower prior in vivo molecular oxidation. CONCLUSIONS: Hyperoxia reduces small bowel injury, accelerates enterocyte turnover, and improves intestinal rehabilitation after IR.


Subject(s)
Enterocytes/physiology , Intestines/blood supply , Oxygen Inhalation Therapy , Oxygen/administration & dosage , Reperfusion Injury/therapy , Animals , Enterocytes/drug effects , Intestinal Mucosa/metabolism , Male , Oxygen/metabolism , Rats , Rats, Sprague-Dawley , Reperfusion Injury/metabolism , Reperfusion Injury/rehabilitation
13.
Am J Physiol Cell Physiol ; 295(1): C2-12, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18434619

ABSTRACT

Infection, simulated by lipopolysaccharide (LPS), is a potent stimulator of tumor necrosis factor-alpha (TNF-alpha) production, and hypoxia often synergizes with LPS to induce higher levels of the secreted cytokine. However, we show that in primary mouse peritoneal macrophages and in three mouse peritoneal macrophage cell lines (RAW 264.7, J774A.1, and PMJ-2R), hypoxia (O(2) < 0.3%) reduces the secretion of LPS-induced TNF-alpha (P < 0.01). In RAW 264.7 cells this reduction was not regulated transcriptionally as TNF-alpha mRNA levels remained unchanged. Rather, hypoxia and LPS reduced the intracellular levels of TNF-alpha by twofold (P < 0.01) by enhancing its degradation in the lysosomes and inhibiting its secretion via secretory lysosomes, as shown by confocal microscopy and verified by the use of the lysosome inhibitor Bafilomycin A1. In addition, although hypoxia did not change the accumulation of the soluble receptor TNF-RII, it increased its binding to the secreted TNF-alpha by twofold (P < 0.05). We suggest that these two posttranslational regulatory checkpoints coexist in hypoxia and may partially explain the reduced secretion and diminished biological activity of TNF-alpha in hypoxic peritoneal macrophages.


Subject(s)
Hypoxia/metabolism , Lysosomes/metabolism , Macrophages, Peritoneal/physiology , Tumor Necrosis Factor-alpha/metabolism , Animals , Cells, Cultured , Female , Lipopolysaccharides/pharmacology , Macrophage Activation , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Protein Processing, Post-Translational , Receptors, Tumor Necrosis Factor, Type II/metabolism
14.
J Leukoc Biol ; 81(6): 1466-76, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17327485

ABSTRACT

Monocytes remodel the extracellular matrix (ECM) by secreting proteins composing the ECM such as fibronectin (FN) and degrading proteases such as matrix metalloproteinase-9 (MMP-9), which cleaves FN into fragments. The effects of FN and its fragmented products on the expression of monocyte MMP-9 are controversial and largely unknown. We showed that in human monocytes, the proinflammatory cytokine TNF-alpha induced MMP-9 secretion and increased fragmentation of FN into distinct fragments. When primary monocytes or the U937 monocytic cell line were incubated on a plastic substrate, plastic-coated with native FN, and plastic-coated with fragmented FN (frag-FN), native FN inhibited TNF-alpha-induced proMMP-9 secretion by twofold (P<0.01) compared with plastic or frag-FN. Exploration of the dynamics of inflammation by incubating cells sequentially on the three substrates showed that frag-FN opposed the inhibitory effect of native FN. Inhibition of proMMP-9 by native FN was exerted at the translational level, as no change in MMP-9 mRNA, intracellular protein accumulation, or proteomic degradation was observed, and when degradation was blocked, no de novo translation of MMP-9 could be measured. We also showed that the reduction of MMP-9 secretion by native FN was responsible for attenuated migration of U937 cells (P<0.05). We suggest that in the inflammatory tissue, intact, native FN has a homeostatic role in harnessing MMP-9 activity. However, as fragmented products accumulate locally, they alleviate the inhibition and enable faster migration of the monocytes through the degraded ECM.


Subject(s)
Fibronectins/metabolism , Matrix Metalloproteinase 9/metabolism , Monocytes/metabolism , Cell Adhesion , Cell Movement , Cells, Cultured , Humans , In Vitro Techniques , Tumor Necrosis Factor-alpha/physiology
15.
Graefes Arch Clin Exp Ophthalmol ; 245(5): 725-32, 2007 May.
Article in English | MEDLINE | ID: mdl-17024442

ABSTRACT

BACKGROUND: The involvement of matrix metalloproteinases (MMPs) in ischemic tissue damage and remodeling has been reported by many investigators. Our study was designed to investigate the involvement of MMPs and of tissue inhibitors of metalloproteinases (TIMPs) in rat retinal ischemic injury, the effect of nitric oxide synthase (NOS) inhibitors on MMPs' activity in this model and whether minocycline (an MMP inhibitor) is protective in retinal ischemia. METHODS: Ninety-four rats were used in the study. Ischemia was induced by 90 min elevation of intraocular pressure. MMPs' activities and the effect of NOS inhibitors [aminoguanidine (AG) or N-nitro-L-arginine (NNA)] and minocycline on MMPs' activities were assessed by zymography and TIMPs expression by Western analysis. Morphological damage was quantified by morphometry of hematoxylin and eosin-stained retinal sections. RESULTS: Retinal extracts exhibited activities of proMMP-9 and proMMP-2. The activity of proMMP-9 increased immediately post ischemia (PI) and peaked to 4.6 times that of normal untreated controls in ischemic retinas and to 2.6 times that of controls in retinas of fellow sham-treated eyes at 24 h PI. The relative amount of TIMP-1 increased to 1.9-fold following ischemia and 2.5-fold in fellow sham-treated eyes at 24 h PI. ProMMP-2 activity increased more than two-fold immediately, at 24 h and at 48 h PI in ischemic retinas, and insignificantly in fellow sham-treated eyes. Treatment with 25 mg/kg AG or NNA caused a non-significant increase in proMMP-9 activity at 24 h PI (3.7- and 2.9-fold, respectively, p>0.6). There was no effect of AG or NNA on the activity of proMMP-2. Minocycline significantly attenuated the retinal ischemic damage, primarily by partially preserving ganglion cells and the inner plexiform layer. Minocyline (0.5 mg/ml or 5 mg/ml) inhibited MMPs' activities in ischemic retinal extracts in vitro. CONCLUSIONS: MMPs participated in morphological ischemic damage to rat retina. Treatment with minocycline dramatically attenuated damage to the retina.


Subject(s)
Ischemia/enzymology , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism , Retinal Diseases/enzymology , Retinal Vessels , Animals , Blotting, Western , Disease Models, Animal , Enzyme Inhibitors/therapeutic use , Guanidines/therapeutic use , Ischemia/pathology , Ischemia/prevention & control , Male , Matrix Metalloproteinase Inhibitors , Minocycline/therapeutic use , Nitric Oxide Synthase/antagonists & inhibitors , Nitroarginine/therapeutic use , Rats , Rats, Sprague-Dawley , Retinal Diseases/pathology , Retinal Diseases/prevention & control , Tissue Inhibitor of Metalloproteinase-1/metabolism
16.
J Leukoc Biol ; 79(4): 706-18, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16434697

ABSTRACT

Cellular hypoxia, characterizing tumors, ischemia, and inflammation induce recruitment of monocytes/macrophages, immobilize them at the hypoxic site, and alter their function. To migrate across the extracellular matrix and as part of their inflammatory functions, monocytes and macrophages secrete proteases, including matrix metalloproteinase-9 (MMP-9), whose expression is induced by proinflammatory cytokines [e.g., tumor necrosis factor alpha (TNF-alpha)]. We show that hypoxia (<0.3% O2 for 48 h) reduced the output of TNF-alpha-induced proMMP-9 by threefold (P < 0.01) in the U937 monocytic cell line and in primary human monocytes. TNF-alpha induced MMP-9 transcription by threefold, but no significant difference was observed in MMP-9 mRNA steady-state between normoxia and hypoxia, which inhibited the trafficking of proMMP-9 via secretory vesicles and increased the intracellular accumulation of proMMP-9 in the cells by 47% and 62% compared with normoxia (P < 0.05), as evaluated by zymography of cellular extracts and confocal microscopy, respectively. Secretion of proMMP-9 was reduced by the addition of cytochalazin B or nocodazole, which inhibits the polymerization of actin and tubulin fibers, or by the addition of the Rho kinase inhibitor Y27632, suggesting the involvement of the cytoskeleton and the Rho GTPases in the process of enzyme secretion. Furthermore, attachment of proMMP-9 to the cell membrane increased after hypoxia via its interactions with surface molecules such as CD44. In addition, the reduced migration of monocytes in hypoxia was shown to be mediated, at least partially, by secreted MMP-9. Thus, hypoxia post-translationally reduced the secreted amounts of proMMP-9 by using two mutually nonexclusive mechanisms: mostly, inhibition of cellular trafficking and to a lesser extent, attachment to the membrane.


Subject(s)
Cell Hypoxia/immunology , Matrix Metalloproteinase 9/drug effects , Matrix Metalloproteinase 9/metabolism , Monocytes/enzymology , Monocytes/metabolism , Binding Sites , Cell Line , Cell Membrane/drug effects , Cell Membrane/immunology , Cell Movement/drug effects , Cell Movement/immunology , Cytochalasin B/pharmacology , Cytokines/antagonists & inhibitors , Cytokines/pharmacology , Dose-Response Relationship, Drug , Humans , Hyaluronan Receptors/immunology , Matrix Metalloproteinase 9/immunology , Monocytes/immunology , Nocodazole/pharmacology , Surface Properties
17.
Am J Physiol Cell Physiol ; 289(5): C1321-31, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16210427

ABSTRACT

Exposure of endothelial cells (ECs) to hypoxia has separately been shown to induce their angiogenesis or death. Matrix metalloproteinase (MMP)-2 is associated with EC angiogenesis, although recent studies also implicate this molecule in EC death. We studied the effect of hypoxia in the absence or presence of TNF-alpha (characteristic of the inflammatory microenvironment accompanying hypoxia) on MMP-2 expression and its role in angiogenesis (proliferation, migration, and tube formation) and in the death of primary human umbilical vein endothelial cells (HUVECs). Hypoxia alone (24-48 h in 0.3% O(2) in the hypoxic chamber) and furthermore, when combined with TNF-alpha, significantly enhanced MMP-2 expression and activity. Hypoxia also led to a reduction in membrane type 1 MMP (MT1-MMP) and tissue inhibitor of metalloproteinase-2 mRNA and protein while enhancing the expression of alpha(v)beta(3) integrin and the cytoskeletal protein phosphopaxillin. Moreover, hypoxia led to colocalization of alpha(v)beta(3) and MMP-2, but not MT1-MMP, with phosphopaxillin in ECs. These results suggest MT1-MMP-independent activation of MMP-2 during hypoxia and support interactions between the ECM, integrins, and the cytoskeleton in hypoxia-induced MMP-2-related functions. Hypoxia enhanced EC migration in an MMP-2-dependent manner while leading to a reduction of cell number via their apoptosis, which was also dependent on MMP-2. In addition, hypoxia caused an aberrant tubelike formation on Matrigel that appeared to be unaffected by MMP-2. The hypoxia-induced, MMP-2-dependent migration of ECs is in accordance with the proangiogenic role ascribed to MMP-2, while the involvement of this protease in the hypoxia-related death of ECs supports an additional apoptotic role for this protease. Hence, in the hypoxic microenvironment, MMP-2 appears to have a dual autocrine role in determining the fate of ECs.


Subject(s)
Cell Death/physiology , Endothelial Cells/physiology , Matrix Metalloproteinase 2/biosynthesis , Oxygen/physiology , Cells, Cultured , Cytoskeletal Proteins/physiology , Gene Expression Regulation/physiology , Humans , Integrin alphaVbeta3/physiology , Matrix Metalloproteinases, Membrane-Associated , Metalloendopeptidases/physiology , Paxillin , Phosphoproteins/physiology , Time Factors , Tissue Inhibitor of Metalloproteinase-2/physiology , Tumor Necrosis Factor-alpha/physiology
18.
Isr Med Assoc J ; 7(6): 351-4, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15984374

ABSTRACT

BACKGROUND: Whereas the diagnosis of classical pertussis has traditionally been based on clinical criteria, increasing numbers of atypical presentations suggest the need for an extensive laboratory-based approach. OBJECTIVES: To assess the relative efficacy of clinical and laboratory methods in the diagnosis of Bordetella pertussis by patient age and immunization status. METHODS: We compared the clinical and laboratory diagnosis of B. pertussis in 87 pre-vaccinated, 78 recently vaccinated, and 75 post-vaccinated children with suspected pertussis. Serum and nasopharyngeal swabs were obtained for serology, culture and polymerase chain reaction. RESULTS: PCR and culture identified 41% and 7% of patients with B. pertussis, respectively (P < 0.001). All positive cultures were PCR-positive. Positive PCR was less common among those recently vaccinated than among those in the pre- (P < 0.001) and post-vaccinated groups (P < 0.05). Positive culture was more common among those pre-vaccinated than among those recently vaccinated (P < 0.01). Positive tests for immunoglobulin M and A were more common among the post-vaccinated than the pre- and recently vaccinated (P < 0.001), respectively. Logistic regression analyses revealed that clinical criteria have no significant association with infection in recently and post-vaccinated children. Among the pre-vaccinated children, whoop and cough duration were associated with a positive PCR (odds ratio 7.66 and 0.5, P < 0.001). Seventy-six percent of pre-vaccinated, 39% of recently vaccinated and 40% of post-vaccinated children with positive PCR did not meet the U.S. Centers for Disease Control diagnostic criteria for B. pertussis. CONCLUSIONS: PCR is a useful tool for pertussis diagnosis, particularly in pre-vaccinated infants. The yield of culture and serology is limited, especially among pre- and recently vaccinated children. In pre-vaccinated infants with whoop and less than 2 weeks of cough, PCR testing should be implemented promptly.


Subject(s)
Bordetella pertussis/isolation & purification , Polymerase Chain Reaction , Whooping Cough/diagnosis , Adolescent , Cells, Cultured/microbiology , Child , Child, Preschool , Enzyme-Linked Immunosorbent Assay , Female , Humans , Infant , Infant, Newborn , Logistic Models , Male , Multivariate Analysis , Predictive Value of Tests , Sensitivity and Specificity , Vaccination , Whooping Cough/immunology
19.
J Neuroimmunol ; 144(1-2): 116-24, 2003 Nov.
Article in English | MEDLINE | ID: mdl-14597105

ABSTRACT

Reciprocal interactions between T cells and antigen-presenting cells (APCs) within the 'Immunological-Synapse' (IS) govern immune cell autoreactivity in multiple sclerosis (MS). The present study examined the expression of a range of co-stimulatory molecules: CD40, CD54, CD80, CD86 and HLA-DR, on the cell-surface of CD14(+) peripheral blood monocytes (PBM) from relapsing-remitting (RR) and secondary-progressive (SP)-MS patients, prior to and during 1 year of Interferon (IFN)-beta-1a (Rebif(R)) therapy. Prior to treatment, patients from both MS subtypes expressed elevated CD80 and reduced CD40 levels in comparison to controls. CD86 expression was significantly reduced in SP compared to RR patients and controls. IFN-beta therapy led to a significant reduction in the expression of CD54 and CD80 in both groups of patients as well as to elevation of CD40 and CD86 expression in SP patients. These results confirm IFN-mediated modulation of the APC surface within the immunological-synapse and implicate CD80 and CD86 as targets for interventional therapies in MS as well as other Th1-mediated autoimmune diseases.


Subject(s)
Adjuvants, Immunologic/therapeutic use , Antigen-Presenting Cells/immunology , Cell Communication/immunology , Interferon-beta/therapeutic use , Multiple Sclerosis, Chronic Progressive/immunology , Multiple Sclerosis, Chronic Progressive/therapy , Multiple Sclerosis, Relapsing-Remitting/immunology , Multiple Sclerosis, Relapsing-Remitting/therapy , Adolescent , Adult , Antigen-Presenting Cells/metabolism , Antigens, CD/biosynthesis , B7-1 Antigen/biosynthesis , B7-2 Antigen , CD40 Antigens/biosynthesis , Down-Regulation/immunology , Female , Flow Cytometry , HLA-DR Antigens/biosynthesis , Humans , Intercellular Adhesion Molecule-1/biosynthesis , Interferon beta-1a , Lipopolysaccharide Receptors/biosynthesis , Male , Membrane Glycoproteins/biosynthesis , Middle Aged , Monocytes/immunology , Monocytes/metabolism , Up-Regulation/immunology
20.
J Immunol ; 171(6): 3225-32, 2003 Sep 15.
Article in English | MEDLINE | ID: mdl-12960352

ABSTRACT

Nitric oxide, produced in macrophages by the high output isoform inducible NO synthase (iNOS), is associated with cytotoxic effects and modulation of Th1 inflammatory/immune responses. Ischemia and reperfusion lead to generation of high NO levels that contribute to irreversible tissue damage. Ischemia and reperfusion, as well as their in vitro simulation by hypoxia and reoxygenation, induce the expression of iNOS in macrophages. However, the molecular regulation of iNOS expression and activity in hypoxia and reoxygenation has hardly been studied. We show in this study that IFN-gamma induced iNOS protein expression (by 50-fold from control, p < 0.01) and nitrite accumulation (71.6 +/- 14 micro M, p < 0.01 relative to control), and that hypoxia inhibited NO production (7.6 +/- 1.7 micro M, p < 0.01) without altering iNOS protein expression. Only prolonged reoxygenation restored NO production, thus ruling out the possibility that lack of oxygen, as a substrate, was the cause of hypoxia-induced iNOS inactivation. Hypoxia did not change the ratio between iNOS monomers and dimers, which are essential for iNOS activity, but the dimers were unable to produce NO, despite the exogenous addition of all cofactors and oxygen. Using immunoprecipitation, mass spectroscopy, and confocal microscopy, we demonstrated in normoxia, but not in hypoxia, an interaction between iNOS and alpha-actinin 4, an adapter protein that anchors enzymes to the actin cytoskeleton. Furthermore, hypoxia caused displacement of iNOS from the submembranal zones. We suggest that the intracellular localization and interactions of iNOS with the cytoskeleton are crucial for its activity, and that hypoxia inactivates iNOS by disrupting these interactions.


Subject(s)
Actinin/metabolism , Macrophages/enzymology , Microfilament Proteins , Nitric Oxide Synthase/antagonists & inhibitors , Nitric Oxide Synthase/metabolism , Actinin/isolation & purification , Animals , Cell Hypoxia/physiology , Cell Line , Cytochalasin B/pharmacology , Dimerization , Down-Regulation/drug effects , Enzyme Activation/physiology , Macrophages/drug effects , Macrophages/metabolism , Mice , Nitric Oxide/metabolism , Nitric Oxide Synthase/biosynthesis , Nitric Oxide Synthase Type II , Nitrites/antagonists & inhibitors , Nitrites/metabolism , Oxygen/metabolism , Spleen/cytology , Spleen/enzymology , Spleen/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL