Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 39
Filter
1.
Sci Rep ; 6: 30088, 2016 07 21.
Article in English | MEDLINE | ID: mdl-27439482

ABSTRACT

A need exists to noninvasively assess renal interstitial fibrosis, a common process to all kidney diseases and predictive of renal prognosis. In this translational study, Magnetic Resonance Imaging (MRI) T1 mapping and a new segmented Diffusion-Weighted Imaging (DWI) technique, for Apparent Diffusion Coefficient (ADC), were first compared to renal fibrosis in two well-controlled animal models to assess detection limits. Validation against biopsy was then performed in 33 kidney allograft recipients (KARs). Predictive MRI indices, ΔT1 and ΔADC (defined as the cortico-medullary differences), were compared to histology. In rats, both T1 and ADC correlated well with fibrosis and inflammation showing a difference between normal and diseased kidneys. In KARs, MRI indices were not sensitive to interstitial inflammation. By contrast, ΔADC outperformed ΔT1 with a stronger negative correlation to fibrosis (R(2) = 0.64 against R(2) = 0.29 p < 0.001). ΔADC tends to negative values in KARs harboring cortical fibrosis of more than 40%. Using a discriminant analysis method, the ΔADC, as a marker to detect such level of fibrosis or higher, led to a specificity and sensitivity of 100% and 71%, respectively. This new index has potential for noninvasive assessment of fibrosis in the clinical setting.


Subject(s)
Fibrosis/diagnosis , Fibrosis/pathology , Image Processing, Computer-Assisted/methods , Kidney Diseases/diagnosis , Kidney Diseases/pathology , Magnetic Resonance Imaging/methods , Adult , Aged , Animals , Female , Histocytochemistry , Humans , Male , Middle Aged , Rats , Sensitivity and Specificity
2.
Arch Mal Coeur Vaiss ; 100(2): 121-5, 2007 Feb.
Article in French | MEDLINE | ID: mdl-17474497

ABSTRACT

The prevalence and characteristics of patients operated for adrenal adenoma (Conn syndrome) as well as their post-operative arterial pressure evolution are varying through literature. Our aim was to report the Grenoble University Hospital experience. From 1993 to 2005, 24 patients (mean age = 46 +/-11 years) presented the biological criteria of primary hyperaldosteronism and benefited from adrenalectomy with confirmation of adrenal adenoma. All had an uncontrolled hypertension, refractory in 42% of cases, with a hypokaliemia (mean = 2.65 +/- 0.47 mmol/l). All adenomas measured more than 10 mm in scanner imaging. After a mean post-operative follow-up of 46 +/- 43 months, 70% of them were normotensive, with (45%) or without (25%) anti-hypertensive therapy. the post-operative kaliemia was normal in all cases. Only 25% had post-operative hormonal dosages for control. Post-operative spontaneous normotensive patients had, at the diagnosis of adrenal adenoma, a more recent and non-refractory hypertension, with a lower number of antihypertensive drugs, a better response to spirinolactone and higher aldosterone plasmatic levels. Two lessons can be taken from this study: 1) Whether 70% of patients operated for adrenal adenoma are normotensive (with or without treatement) post-operatively, only 25% are definitely cured after 4 years. Factors associated to a post-operative cure highlight the interest of an ealy diagnosis. 2) There is probably an underdiagnosis of adrenal adenoma (Conn syndrome) because neither adenomas with normokaliemia, nor adenomas <10 mm in scanner imaging have ever been diagnosed or at least, sent to surgery.


Subject(s)
Adrenal Cortex Neoplasms/diagnosis , Adrenal Cortex Neoplasms/surgery , Adrenocortical Adenoma/diagnosis , Adrenocortical Adenoma/surgery , Adrenal Cortex Neoplasms/complications , Adrenalectomy , Adrenocortical Adenoma/complications , Adult , Blood Pressure , Female , Follow-Up Studies , Humans , Hyperaldosteronism/etiology , Male , Middle Aged , Prognosis , Retrospective Studies
3.
Apoptosis ; 7(2): 91-8, 2002 Apr.
Article in English | MEDLINE | ID: mdl-11865192

ABSTRACT

Infection of mice with Plasmodium Berghei Anka (PbA) leads to a thrombocytopenia, due to a reduced platelet life span, eventually associated with a syndrome of severe or cerebral malaria (CM). Thrombocytopenia was associated with an increase in the number of microparticles (mcp) in plasma. More than >60% of these mcp were of platelet origin, as seen by staining with an anti-platelet antibody. The thrombocytopenia and the amount of mcp were decreased in mice treated with anti CD40L mAb, suggesting that CD40L is the main effector of the thrombocytopenia. Caspase-1, -3, -6, -8, -9 were activated in platelets from infected mice, as seen by the binding of labeled probes or the amount of pro-caspase-3. Treatment of infected mice with the caspases inhibitor ZVAD-fmk decreased the number of mcp and the thrombocytopenia, showing that platelet caspases are responsible for platelet fragmentation. In addition, the caspase inhibitor also caused a decrease in the mortality associated with CM, indicating a critical role of caspases in the expression of CM.


Subject(s)
Blood Platelets/pathology , Malaria/enzymology , Thrombocytopenia/enzymology , Amino Acid Chloromethyl Ketones/pharmacology , Animals , Blood Platelets/cytology , Blotting, Western , CD40 Ligand/biosynthesis , Caspase 1/metabolism , Caspase 3 , Caspase 6 , Caspase 8 , Caspase 9 , Caspases/metabolism , Cricetinae , Cysteine Proteinase Inhibitors/pharmacology , Disease Models, Animal , Enzyme Activation , Flow Cytometry , Megakaryocytes/metabolism , Mice , Mice, Inbred C57BL , Microscopy, Fluorescence , Peptides/chemistry , Plasmodium berghei/pathogenicity , Protein Binding , Time Factors , Tumor Necrosis Factor-alpha/metabolism
4.
Am J Pathol ; 159(2): 733-42, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11485931

ABSTRACT

We explored the role of CD40-CD40L (CD154) in the severe malaria elicited by Plasmodium berghei anka infection in mice. Mortality was >90% by day 8 after infection in +/+ mice, but markedly decreased in CD40-/- or in CD40L-/- mice, as well as in +/+ mice treated with anti-CD40L monoclonal antibody. Parasitemia was similar in the different conditions. Breakdown of the blood-brain barrier was evident in infected +/+, but not in CD40-/- mice. Thrombocytopenia was less severe in CD40-/- mice than in the +/+ controls. Sequestration of macrophages in brain venules and alveolar capillaries was reduced in CD40-/- or in CD40L-/- mice, whereas sequestration of parasitized red blood cells or polymorphonuclear leukocytes in alveolar capillaries was CD40-CD40L-independent. CD40 mRNA was increased in the brain and lung of infected mice whereas CD40L was increased in the lung. Tumor necrosis factor plasma levels were similarly increased in infected +/+ or CD40-/- mice. Expression of CD54 and its mRNA levels in the brain were moderately decreased in CD40-deficient mice. Thus the mortality associated with severe malaria requires CD40-CD40L interaction that contributes to the breakdown of the blood-brain barrier, macrophage sequestration, and platelet consumption.


Subject(s)
CD40 Antigens/physiology , CD40 Ligand/physiology , Malaria/immunology , Plasmodium berghei , Animals , Blood-Brain Barrier , Brain/immunology , Brain/pathology , CD40 Antigens/genetics , CD40 Ligand/genetics , Gene Expression Regulation/immunology , Intercellular Adhesion Molecule-1/genetics , Macrophages/physiology , Malaria/blood , Malaria/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Platelet Count , RNA, Messenger/genetics , Thrombocytopenia , Transcription, Genetic , Tumor Necrosis Factor-alpha/metabolism
5.
Eur J Cell Biol ; 80(2): 171-7, 2001 Feb.
Article in English | MEDLINE | ID: mdl-11302522

ABSTRACT

We explored the role of CD18 (beta2 integrin) in platelet physiology, using mice genetically deficient in CD18 (CD18 -/-), or its main ligand CD54 (ICAM-1, CD54 -/-). CD18 and CD11a were evident in platelets from +/+, but not from CD18 -/- mice, as seen by immunofluorescence or Western blots. CD18 mRNA was also detectable by RT-PCR in platelets from +/+, but not from CD18 -/- mice. The life span of platelets was significantly shorter in CD18 -/- than in +/+ or CD54 -/- mice, as seen by in vivo biotinylation. When a local inflammation was elicited by the intra-tracheal injection of TNF, labeled platelets from +/+, but not from CD18 -/- donors, did localize in the lung. The content of Bcl-3 was about 20-fold higher in platelet from CD18 -/-, than in those from +/+ or CD54 -/- donors, as seen on Western blots or by immunofluorescence and flow cytometry, while the amount of pro-caspase-3 was decreased. An activation of caspases in platelets from CD18 -/- was also evidenced by protease assays. Accordingly, gelsolin, a protein cleaved by caspase-3, showed a low-molecular-weight band in platelets from CD18 -/- but not from +/+ donors. These results demonstrate that the beta2 integrin, present in mouse platelets, modulates caspase activation and consequently platelet life span and response to TNF.


Subject(s)
Blood Platelets/metabolism , CD18 Antigens/metabolism , Caspases/metabolism , Animals , Annexin A5/metabolism , B-Cell Lymphoma 3 Protein , Blood Platelets/cytology , Blood Platelets/physiology , Blotting, Western , CD18 Antigens/biosynthesis , CD18 Antigens/genetics , Caspase 3 , Cell Adhesion , Enzyme Activation , Gelsolin/metabolism , Gene Expression , Intercellular Adhesion Molecule-1/genetics , Intercellular Adhesion Molecule-1/physiology , Lymphocyte Function-Associated Antigen-1/biosynthesis , Mice , Mice, Inbred C57BL , Mice, Knockout , Proto-Oncogene Proteins/metabolism , RNA, Messenger , Reverse Transcriptase Polymerase Chain Reaction , Transcription Factors , Tumor Necrosis Factor-alpha/pharmacology
6.
Dev Immunol ; 8(3-4): 183-91, 2001.
Article in English | MEDLINE | ID: mdl-11785668

ABSTRACT

Incidence of apoptosis was investigated in the spleen and lymph nodes of +/+, CD18 -/- and urokinase receptor (uPAR, CD87) -/- mice, untreated or Plasmodium Berghei Anka (PbA) infected. In non infected mice, incidence of apoptosis was lower in the lymph nodes of CD18 -/- and uPAR -/- than in +/+ mice, as seen by FACS analysis to count the number of hypodiploid and Annexin-V binding cells. Infection of mice with PbA resulted in a marked increase in the size of spleen and lymph nodes 7-8 days after infection, which was slightly higher in uPAR -/- and CD 18 -/- than in +/+ mice. PbA infection increased about 7 fold the incidence of apoptosis in the lymphoid organs of +/+, especially in the white pulp and germinal centers of the spleen and lymph nodes, while in contrast it was unchanged in PbA infected CD 18 -/- or uPAR -/- mice. Serum IgG levels, and number of circulating leukocytes were significantly higher in both uPAR and CD18 -/- than in +/+ mice. These results indicate that the CD18 and uPAR surface molecules, which are known to be associated in the cell membrane, have an important influence upon the incidence of cell survival in both normal or stimulated lymphoid organs.


Subject(s)
Apoptosis , CD18 Antigens/physiology , Lymphoid Tissue/cytology , Lymphoid Tissue/pathology , Malaria/immunology , Receptors, Cell Surface/physiology , Animals , Annexin A5/analysis , CD18 Antigens/genetics , Female , Immunoglobulin G/blood , Leukocyte Count , Lymph Nodes/cytology , Lymph Nodes/pathology , Lymphokines , Malaria/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Plasmodium berghei/pathogenicity , Receptors, Cell Surface/genetics , Receptors, Urokinase Plasminogen Activator , Spleen/cytology , Spleen/pathology , Splenomegaly/pathology
7.
Eur Cytokine Netw ; 11(4): 662-8, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11125311

ABSTRACT

Lipopolysaccharide (LPS)-induced effects on energy balance are characterized by alterations in energy expenditure (hypermetabolism) and food intake (anorexia). To study the role of tumour necrosis factor alpha (TNF-alpha) on some of these metabolic responses to endotoxin, we have used transgenic mice expressing soluble tumour necrosis factor receptor-1 IgG fusion protein (TNFR1-IgG) as well as TNF-alpha knockout (KO), lymphotoxin-alpha (LT-alpha) KO, and interferon-gamma receptor (IFN-gamma R) KO mice. The results from TNFR1-IgG transgenic mice suggest that the hypermetabolic and anorectic responses induced by LPS are independently regulated since, in the absence of TNF-alpha or LT-alpha, the LPS-induced hypermetabolism is almost prevented but not the anorexia. The anorectic response shows the strongest association with IFN-gamma since both IFN-gamma R KO mice and mice treated with anti-IFN-gamma antibody showed marked reduction in the LPS-induced anorexia compared to other mice. IFN-gamma R KO mice also have an attenuated thermogenic response to endotoxin. Anti-Asialo GM1 antibody treatment attenuated both the hypermetabolic and anorectic responses to LPS, to an extent comparable to that observed in IFN-gamma R KO mice. This finding suggests that natural killer cells (lymphocytic subsets) may be involved in IFN-gamma production and play an important role in the metabolic alterations induced by LPS. We also showed that the hypermetabolic response of control mice is associated with an upregulation of cytokine expression within the brain and an increase in permeability of the blood brain barrier. LPS-induced anorexia appears to involve peripheral cytokine expression.


Subject(s)
Eating , Energy Metabolism , Interferon-gamma/immunology , Lipopolysaccharides/pharmacology , Tumor Necrosis Factor-alpha/immunology , Animals , Antigens, CD/genetics , Blood-Brain Barrier/drug effects , Brain/anatomy & histology , Brain/drug effects , Brain/immunology , Cytokines/biosynthesis , Cytokines/blood , Cytokines/genetics , Energy Metabolism/drug effects , Evans Blue/metabolism , Female , Interferon-gamma/biosynthesis , Lymphotoxin-alpha/genetics , Lymphotoxin-alpha/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , RNA, Messenger/biosynthesis , Receptors, Interferon/genetics , Receptors, Tumor Necrosis Factor/genetics , Receptors, Tumor Necrosis Factor, Type I , Spleen/drug effects , Spleen/immunology , Transcriptional Activation , Tumor Necrosis Factor-alpha/biosynthesis , Tumor Necrosis Factor-alpha/genetics , Interferon gamma Receptor
8.
Infect Immun ; 68(7): 3822-9, 2000 Jul.
Article in English | MEDLINE | ID: mdl-10858190

ABSTRACT

We explored the role of urokinase and tissue-type plasminogen activators (uPA and tPA), as well as the uPA receptor (uPAR; CD87) in mouse severe malaria (SM), using genetically deficient (-/-) mice. The mortality resulting from Plasmodium berghei ANKA infection was delayed in uPA(-/-) and uPAR(-/-) mice but was similar to that of the wild type (+/+) in tPA(-/-) mice. Parasitemia levels were similar in uPA(-/-), uPAR(-/-), and +/+ mice. Production of tumor necrosis factor, as judged from the plasma level and the mRNA levels in brain and lung, was markedly increased by infection in both +/+ and uPAR(-/-) mice. Breakdown of the blood-brain barrier, as evidenced by the leakage of Evans Blue, was similar in +/+ and uPAR(-/-) mice. SM was associated with a profound thrombocytopenia, which was attenuated in uPA(-/-) and uPAR(-/-) mice. Administration of aprotinin, a plasmin antagonist, also delayed mortality and attenuated thrombocytopenia. Platelet trapping in cerebral venules or alveolar capillaries was evident in +/+ mice but absent in uPAR(-/-) mice. In contrast, macrophage sequestration in cerebral venules or alveolar capillaries was evident in both +/+ and uPAR(-/-) mice. Polymorphonuclear leukocyte sequestration in alveolar capillaries was similar in +/+ and uPAR(-/-) mice. These results demonstrate that the uPAR deficiency attenuates the severity of SM, probably by its important role in platelet kinetics and trapping. These results therefore suggest that platelet sequestration contributes to the pathogenesis of SM.


Subject(s)
Malaria/metabolism , Plasmodium berghei , Receptors, Cell Surface/deficiency , Urokinase-Type Plasminogen Activator/deficiency , Animals , Aprotinin/pharmacology , Blood Platelets/pathology , Blood-Brain Barrier , Cell Survival , Fibrinogen/metabolism , Kinetics , Lung/pathology , Malaria/complications , Malaria/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Organ Size , Parasitemia/complications , Parasitemia/genetics , Parasitemia/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Cell Surface/genetics , Receptors, Urokinase Plasminogen Activator , Spleen/pathology , Thrombocytopenia/etiology , Tissue Plasminogen Activator/deficiency , Tissue Plasminogen Activator/genetics , Tumor Necrosis Factor-alpha/genetics , Urokinase-Type Plasminogen Activator/genetics
9.
Hematol J ; 1(3): 199-205, 2000.
Article in English | MEDLINE | ID: mdl-11920190

ABSTRACT

INTRODUCTION: Plasminogen activators (PA) and plasmin are known to affect platelets but little is known of their role in platelet kinetics. We took advantage of genetically deficient mice to explore the role of urokinase (uPA) and tissue type (tPA) PAs, as well as the uPA receptor (uPAR, CD87) in platelet kinetics. MATERIALS AND METHODS: Platelet shape and number were investigated by flow cytometry. Platelet kinetics was investigated by the in vivo biotinylation and FACS analysis. Platelet production was investigated by counting megakaryocytes in bone marrow. RESULTS: Platelets counts were within the same range in wild type (+/+), uPA, tPA and uPAR-deficient mice. Platelet survival was similar in +/+, uPA-/-, tPA-/- but markedly reduced in uPAR-/- mice. The number of megakaryocytes in bone marrow and spleen was increased 2-3-fold in uPAR-/- compared to +/+ mice. TGF-beta mRNA level within the bone marrow was also significantly increased in uPAR-/- mice. Consistent with an increased platelet production, platelets from uPAR-/- mice had a higher RNA content, as seen by Propidium Iodide (PI) labeling and FACS analysis. Since uPAR is detectable in both hemopoietic and non-hemopoietic cells, radiation chimera were prepared. Investigation of platelet kinetics in chimera showed that platelet survival is reduced with a deficit in either bone marrow-derived, or non-hemopoietic, host cells. CONCLUSION: These results demonstrate that uPAR, but not uPA or tPA, is essential for maintaining normal platelet survival. In addition, uPAR-/- mice maintain normal platelet numbers through increased production.


Subject(s)
Blood Platelets/physiology , Plasminogen Activators/blood , Receptors, Cell Surface/blood , Animals , Fibrinolysin/metabolism , Kinetics , Megakaryocytes/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Cell Surface/deficiency , Receptors, Cell Surface/genetics , Receptors, Urokinase Plasminogen Activator , Tissue Plasminogen Activator/blood
10.
Circulation ; 99(25): 3315-21, 1999 Jun 29.
Article in English | MEDLINE | ID: mdl-10385508

ABSTRACT

BACKGROUND: Urokinase plasminogen activator receptor (uPAR, CD87) is a widely distributed 55-kD, glycoprotein I-anchored surface receptor. On binding of its ligand uPA, it is known to increase leukocyte adhesion and traffic. Using genetically deficient mice, we explored the role of uPAR in platelet kinetics and TNF-induced platelet consumption. METHODS AND RESULTS: Anti-uPAR antibody stained platelets from normal (+/+) but not from uPAR-/- mice, as seen by fluorescence-activated cell sorter analysis. 51Cr-labeled platelets from uPAR-/- donors survived longer than those from +/+ donors when injected into a +/+ recipient. Intratracheal TNF injection induced thrombocytopenia and a platelet pulmonary localization, pronounced in +/+ but absent in uPAR-/- mice. Aprotinin, a plasmin inhibitor, decreased TNF-induced thrombocytopenia. TNF injection markedly reduced the survival and increased the pulmonary localization of 51Cr-labeled platelets from +/+ but not from uPAR-/- donors, indicating that it is the platelet uPAR that is critical for their response to TNF. As seen by electron microscopy, TNF injection increased the number of platelets and polymorphonuclear neutrophils (PMNs) in the alveolar capillaries of +/+ mice, whereas in uPAR-/- mice, platelet trapping was insignificant and PMN trapping was slightly reduced. Platelets within alveolar capillaries of TNF-injected mice were activated, as judged from their shape, and this was evident in +/+ but not in uPAR-/- mice. CONCLUSIONS: These results demonstrate for the first time the critical role of platelet uPAR for kinetics as well as for activation and endothelium adhesion associated with inflammation.


Subject(s)
Endothelium, Vascular/metabolism , Plasminogen Activators/metabolism , Receptors, Cell Surface/metabolism , Tumor Necrosis Factor-alpha/metabolism , Animals , Aprotinin/pharmacology , Capillaries , Cell Adhesion , Cell Survival , Chromium Radioisotopes , Endothelium, Vascular/cytology , Flow Cytometry , Injections , Kinetics , Mice , Mice, Inbred C57BL , Neutrophils/metabolism , Plasminogen Activators/pharmacology , Pulmonary Alveoli/blood supply , Receptors, Urokinase Plasminogen Activator , Recombinant Proteins/metabolism , Recombinant Proteins/pharmacology , Thrombocytopenia/metabolism , Thrombocytopenia/prevention & control , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Tumor Necrosis Factor-alpha/pharmacology
11.
Am J Pathol ; 154(5): 1479-87, 1999 May.
Article in English | MEDLINE | ID: mdl-10329601

ABSTRACT

Keratinocyte growth factor (KGF) has been used successfully to prevent alveolar damage induced by oxygen exposure in rodents. However, this treatment was used intratracheally and before oxygen exposure, which limited its clinical application. In the present study, mice were treated with the recombinant human KGF intravenously before (days -2 and -1) or during (days 0 and +1) oxygen exposure. In both cases, lung damage was attenuated. KGF increased the number of cells incorporating bromodeoxyuridine (BrdU) in the septa and in bronchial epithelium of air-breathing mice but not of oxygen-exposed mice, indicating that the protective effect of KGF is not necessarily associated with proliferation. Oxygen-induced damage of alveolar epithelium and, unexpectedly, of endothelium was prevented by KGF treatment as seen by electron microscopy. We investigated the effect of KGF on different mechanisms known to be involved in oxygen toxicity. The induction of p53, Bax, and Bcl-x mRNAs during hyperoxia was to a large extent prevented by KGF. Surfactant proteins A and B mRNAs were not markedly modified by KGF. The anti-fibrinolytic activity observed in the alveoli during hyperoxia was to a large extent prevented by KGF, most probably by suppressing the expression of plasminogen activator inhibitor-1 (PAI-1) mRNA and protein. As PAI-1 -/- mice are more resistant to hyperoxia, KGF might act, at least in part, by decreasing the expression of this protease inhibitor and by restoring the fibrinolytic activity into the lungs.


Subject(s)
Endothelium, Vascular/drug effects , Fibroblast Growth Factors , Growth Substances/therapeutic use , Hyperoxia/pathology , Keratinocytes , Oxygen/antagonists & inhibitors , Pulmonary Alveoli/drug effects , Animals , Cell Division/drug effects , Cell Size/drug effects , DNA Fragmentation , Drug Evaluation, Preclinical , Epithelium/drug effects , Female , Fibroblast Growth Factor 10 , Fibroblast Growth Factor 7 , Mice , Mice, Inbred C57BL , Mice, Inbred CBA , Microscopy, Electron , RNA, Messenger/biosynthesis
12.
Lab Invest ; 79(4): 495-500, 1999 Apr.
Article in English | MEDLINE | ID: mdl-10212002

ABSTRACT

Injection of mouse recombinant TNF to mice induced apoptosis and detachment of the enterocytes of the tip of the villi, evident after 30 to 90 minutes, which resulted in a shrinkage of the villi. Injection of TNF increased the expression of caspase 1, 2, 3, and 6 as well as of cathepsin D in the mucosal wall, which was maximal 30 minutes after TNF injection. Caspase 1 and 3 were not induced in TNFR1-deficient mice in which TNF does not induce apoptosis and detachment. The administration of a caspase inhibitor (ZVAD-fmk, 300 microg) decreased enterocyte detachment and apoptosis, as well as villus atrophy, whereas a caspase 3 inhibitor (Z-DEVD-cmk) had no effect. The results indicate that the induction of caspases by TNF is the cause of their detachment in the lumen and of the resulting villus atrophy.


Subject(s)
Amino Acid Chloromethyl Ketones/pharmacology , Apoptosis/physiology , Cell Adhesion/physiology , Intestinal Mucosa/cytology , Tumor Necrosis Factor-alpha/pharmacology , Animals , Antigens, CD/genetics , Antigens, CD/metabolism , Antigens, CD/physiology , Apoptosis/drug effects , Cell Adhesion/drug effects , Cysteine Proteinase Inhibitors/pharmacology , DNA Fragmentation , Intestinal Mucosa/drug effects , Intestinal Mucosa/pathology , Mice , Mice, Inbred C57BL , Mice, Inbred CBA , Mice, Knockout , Receptors, Tumor Necrosis Factor/genetics , Receptors, Tumor Necrosis Factor/metabolism , Receptors, Tumor Necrosis Factor/physiology , Receptors, Tumor Necrosis Factor, Type I , Recombinant Proteins/pharmacology
13.
Eur J Immunol ; 28(11): 3499-505, 1998 11.
Article in English | MEDLINE | ID: mdl-9842892

ABSTRACT

Injection of recombinant mouse TNF into mice is known to induce a shrinkage of the duodenal villi, which becomes evident 30-90 min later and is associated with a detachment of enterocytes in the lumen. These cells can be collected by lavage and are all apoptotic, i.e. hypodiploid as seen by flow cytometric analysis. Thus the count of detached cells was used as an evaluation of the TNF-induced cell loss and apoptosis in the mucosa. TNF injection induced a cell loss of similar magnitude in wild-type (+/+) or in mice lacking the TNF receptor (TNFR)2 (p75, TNFR2-/-), while mice lacking the TNFR1 (p55, TNFR1-/-) were completely resistant to this effect. TNF increased the expression of p53 tumor suppressor gene in the enterocytes from the crypts but not from the villi, as seen by Western blots and histochemistry. TNF increased the expression of p53 in both TNFR2-/- and TNFR1-/- mice. Furthermore, enterocyte cell loss was not attenuated in p53-/- mice. The results indicate that TNF, acting on its receptor 1, induces an apoptotic detachment of the enterocytes from the tip of the villi (i.e. the old enterocytes), while in the enterocytes from the crypts (the young enterocytes) TNF increases, via either TNFR1 or TNFR2, the expression of p53, without inducing apoptosis.


Subject(s)
Apoptosis/drug effects , Intestinal Mucosa/drug effects , Receptors, Tumor Necrosis Factor/physiology , Tumor Necrosis Factor-alpha/pharmacology , Tumor Suppressor Protein p53/physiology , Animals , Intestinal Mucosa/pathology , Mice , Mice, Inbred C57BL , Mice, Inbred CBA
14.
Immunology ; 95(1): 111-6, 1998 Sep.
Article in English | MEDLINE | ID: mdl-9767465

ABSTRACT

We explored the thrombocytopaenia elicited by the i.v. injection of mouse recombinant tumour necrosis factor (TNF) in mice. Injection of 10 micrograms of TNF led to a thrombocytopaenia (evident after 0.5 hr) which was caused by decreased platelet survival, as seen by the injection of labelled platelets. TNF-induced thrombocytopaenia was not prevented by heparin, nor by depletion of either fibrinogen or C'. TNF-induced thrombocytopaenia was markedly attenuated in mice treated with reserpine, an agent that depletes monoamines from mast cells and other cells, and in the mast-cell-deficient WWv mice. In vitro, TNF elicited a modest release of monoamine from peritoneal mast cells and from a mast cell line. When mice are injected with 3H-serotonin (3H-5HT) before TNF, TNF injection increased the plasma 3H-5HT content 1 hr later, modifications absent in reserpine pretreated or mast-cell-deficient mice. 3H-5HT content of the small intestine was markedly depleted in TNF-injected mice, suggesting that this organ is the source of the plasma 3H-5HT. Drop in body temperature and mortality induced by TNF were also attenuated in mast-cell-deficient, and in reserpine pretreated mice. These results indicate that TNF can induce a release of monoamines from mast cells, mainly from those of the small intestine, a process that contributes to TNF-induced thrombocytopaenia and mortality.


Subject(s)
Mast Cells/metabolism , Serotonin/metabolism , Thrombocytopenia/immunology , Tumor Necrosis Factor-alpha/pharmacology , Adrenergic Uptake Inhibitors/therapeutic use , Animals , Body Temperature/drug effects , Cell Line , Cells, Cultured , Injections, Intravenous , Intestine, Small/metabolism , Mast Cells/drug effects , Mice , Mice, Inbred C57BL , Mice, Inbred CBA , Mice, Mutant Strains , Platelet Count/drug effects , Recombinant Proteins/pharmacology , Reserpine/therapeutic use , Thrombocytopenia/metabolism , Thrombocytopenia/mortality , Tritium
15.
J Immunol ; 160(12): 6182-6, 1998 Jun 15.
Article in English | MEDLINE | ID: mdl-9637537

ABSTRACT

An injection of TNF in mice induced profound thrombocytopenia, due to an increase of platelet consumption, that was evident after 1 h and lasted for 3 days. This process was evident in mice that were genetically deficient in TNFR2 (p75) but not in mice lacking TNFR1 (p55), indicating that the process is mediated by TNFR1-bearing cells. To explore the site of action of TNF, labeled platelets from TNFR1 -/- or +/+ donors were transferred to TNFR1 -/- or +/+ recipients. TNF induced the consumption of platelets from TNFR1 -/- donors when injected into +/+ recipients, while platelets from +/+ donors were not consumed when present in TNFR1 -/- recipients; this finding indicates that TNF acts on the TNFR1 of host cells but does not act on platelets. The expression of TNFRs is consistent with this interpretation, since TNFRs were not detected on platelets by flow cytometry. In megakaryocytes, the expression of TNFR1 was detected by immunohistochemistry. These results indicate that TNF induces platelet consumption by acting not on platelets directly but on the TNFR1 of other cells, presumably increasing the release of factors with agonist activity for platelets.


Subject(s)
Antigens, CD/physiology , Receptors, Tumor Necrosis Factor/physiology , Thrombocytopenia/immunology , Tumor Necrosis Factor-alpha , Animals , Blood Platelets/drug effects , Humans , Immunohistochemistry , Mice , Mice, Inbred C57BL , Mice, Inbred CBA , Mice, Knockout , Receptors, Tumor Necrosis Factor, Type I , Receptors, Tumor Necrosis Factor, Type II , Thrombocytopenia/chemically induced
16.
Am J Pathol ; 151(5): 1397-405, 1997 Nov.
Article in English | MEDLINE | ID: mdl-9358766

ABSTRACT

Tumor necrosis factor-alpha (TNF) is known to be an important mediator in the pathogenesis of several inflammatory diseases. Vascular endothelial cells represent a major target of TNF effects. Platelet sequestration has been found in brain microvessels during experimental cerebral malaria and lung in experimental pulmonary fibrosis, implying that it may participate in TNF-dependent microvascular pathology. In this study, we investigated the mechanisms of platelet-endothelial interaction, using co-cultures between platelets and TNF-activated mouse brain microvascular endothelial cells (MVECs). Adhesion and fusion of platelets to MVECs was evidenced by electron microscopy, dye transfer, and flow cytometry. It was induced by TNF and interferon-gamma and depended on LFA-1 expressed on the platelet surface and ICAM-1 expressed on MVECs. The adhesion and fusion also led to the transfer of platelet markers on the MVEC surface, rendering these more adherent for leukocytes, and to an enhanced MVEC sensitivity to TNF-induced injury. These results suggest that platelets can participate in TNF-induced microvascular pathology.


Subject(s)
Blood Platelets/physiology , Cerebrovascular Circulation/drug effects , Endothelium, Vascular/drug effects , Endothelium, Vascular/pathology , Tumor Necrosis Factor-alpha/pharmacology , Animals , Cell Fusion/physiology , Cells, Cultured , Coculture Techniques , Endothelium, Vascular/physiopathology , Female , Mice , Mice, Inbred CBA , Microcirculation/drug effects , Microscopy, Electron , Platelet Adhesiveness/physiology
17.
Am J Respir Cell Mol Biol ; 15(1): 107-14, 1996 Jul.
Article in English | MEDLINE | ID: mdl-8679214

ABSTRACT

Mice were exposed to pure oxygen for various times to explore the pulmonary platelet trapping associated with alveolar damage, its mechanism, and its role in the lesions. Platelet sequestration, evaluated by electron microscopy and by injection of radiolabeled platelets, was detectable after 72 h and reached a maximum after 96 h of exposure (i.e., shortly before death). Circulating platelets (analyzed by Facscan) showed some increase in the expression of CD11a and CD62, but little change in CD31 and CD61. Both platelet activation and lung sequestration were dependent on TNF-alpha, since antibody against TNF-alpha reduced the expression of CD11a on circulating platelets and their sequestration in the lung. Lung platelet sequestration was also decreased by anti-CD11a MoAb. Northern blot analysis of lung mRNA isolated at 96 h of oxygen exposure revealed a 7-fold increase in CD54 (intercellular adhesion molecule-1 [ICAM-1]) and a 2.5-fold increase in TNF-alpha mRNAs respectively. These results demonstrate that the platelet pulmonary trapping induced by hyperoxia is dependent upon TNF-alpha and the CD11a-CD54 adhesion molecules. However, platelet trapping does not appear to play an important pathogenic role in acute oxygen injury, since treatments that decrease trapping (anti-TNF-alpha, anti-CD11a, or antibody-induced thrombocytopenia) did not markedly attenuate the alveolar damage.


Subject(s)
Bronchopulmonary Sequestration/etiology , Hyperoxia/complications , Platelet Activation/physiology , Animals , Antibodies, Monoclonal , CD18 Antigens/immunology , Flow Cytometry , Hyperoxia/physiopathology , Intercellular Adhesion Molecule-1/genetics , Mice , Mice, Inbred C57BL , Mice, Inbred CBA , Neutrophils/physiology , Oxygen/toxicity , Pulmonary Alveoli/cytology , Pulmonary Alveoli/immunology , Pulmonary Alveoli/physiopathology , RNA, Messenger/analysis , Thrombocytopenia/physiopathology , Time Factors , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/immunology
18.
Eur J Haematol ; 56(3): 130-7, 1996 Mar.
Article in English | MEDLINE | ID: mdl-8598231

ABSTRACT

The expression of molecules of the beta2 integrin family (CD11a, CD11b , CD11c, and CD18) was explored on 2 human megakaryocytic cell lines and on platelets from different donors by immunofluorescence and flow cytometry using a large panel of mAb. CD11a, CD11b, CD11c and CD18 were detected on the megakaryocytic cell lines DAMI and HEL. A low and variable expression of CD11a, CD11b and CD18 determinants was also detected on resting platelets; this expression was markedly increased when platelets were activated by thrombin. Expression of CD18 was closely correlated to that of CD11a or CD11b when comparing the fluorescence intensity observed in different experiments. In presence of Ca++, platelets did bind to a RAJI cell line which exhibits a high expression level of CD54. This binding was increased when platelets were activated by thrombin and was decreased by an anti CD11a, CD18 and anti CD54 mAb. This study indicates that human platelets express molecules of the beta2 integrin family, when activated, which allows them to bind to CD54 bearing cells.


Subject(s)
Blood Platelets/metabolism , CD11 Antigens/biosynthesis , CD18 Antigens/biosynthesis , Platelet Activation , Animals , Antigens, Neoplasm/metabolism , Blood Platelets/drug effects , Burkitt Lymphoma/pathology , CD11 Antigens/genetics , CD18 Antigens/genetics , Calcium/pharmacology , Culture Media/pharmacology , Gene Expression Regulation , Humans , Intercellular Adhesion Molecule-1/metabolism , Leukemia, Erythroblastic, Acute/pathology , Mice , Platelet Activation/drug effects , Species Specificity , Thrombin/pharmacology , Tumor Cells, Cultured
19.
J Clin Invest ; 96(1): 250-9, 1995 Jul.
Article in English | MEDLINE | ID: mdl-7542280

ABSTRACT

The murine TNF-alpha gene was expressed under the control of the human surfactant protein SP-C promoter in transgenic mice. A number of the SP-C TNF-alpha mice died at birth or after a few weeks with very severe lung lesions. Surviving mice transmitted a pulmonary disease to their offspring, the severity and evolution of which was related to the level of TNF-alpha mRNA in the lung; TNF-alpha RNA was detected in alveolar epithelium, presumably in type II epithelial cells. In a longitudinal study of two independent mouse lines, pulmonary pathology, at 1-2 mo of age, consisted of a leukocytic alveolitis with a predominance of T lymphocytes. Leukocyte infiltration was associated with endothelial changes and increased levels of mRNA for the endothelial adhesion molecule VCAM-1. In the following months, alveolar spaces enlarged in association with thickening of the alveolar walls due to an accumulation of desmin-containing fibroblasts, collagen fibers, and lymphocytes. Alveolar surfaces were lined by regenerating type II epithelial cells, and alveolar spaces contained desquamating epithelial cells in places. Platelet trapping in the damaged alveolar capillaries was observed. Pulmonary pathology in the SP-C TNF-alpha mice bears a striking resemblance to human idiopathic pulmonary fibrosis, in which increased expression of TNF-alpha in type II epithelial cells has also been noted. These mice provide a valuable animal model for understanding the pathogenesis of pulmonary fibrosis and exploring possible therapeutic approaches.


Subject(s)
Disease Models, Animal , Pulmonary Fibrosis/etiology , Tumor Necrosis Factor-alpha/genetics , Animals , Base Sequence , Cell Adhesion Molecules/genetics , Female , Immunohistochemistry , Lung/pathology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred DBA , Mice, Transgenic , Molecular Sequence Data , RNA, Messenger/analysis , Transforming Growth Factor beta/genetics , Vascular Cell Adhesion Molecule-1
20.
Exp Lung Res ; 21(2): 227-37, 1995.
Article in English | MEDLINE | ID: mdl-7539740

ABSTRACT

The role of bombesin was investigated in the course of the pulmonary inflammation and fibrosis (PF) elicited by the intratracheal (IT) or intravenous (i.v.) administration of bleomycin in mice. Bleomycin-induced alveolitis was associated with an accumulation of cells, presumably macrophages, containing bombesin, as evidenced by immunohistochemistry. Administration of bombesin by an osmotic minipump implanted IP, at a rate of 6 micrograms/h, decreased the lung hydroxyproline evident 15 days after IV or IT administration of bleomycin. In contrast, antibombesin monoclonal antibody 2A11 (mAb) increased the lung hydroxyproline content after IT administration of bleomycin. In addition, the mortality of bleomycin-injected mice was increased by the anti-bombesin mAb. Bombesin administration induced an increase, and anti-bombesin mAb induced a decrease, in the number of macrophages recovered from the bronchoalveolar lavage. Administration of bombesin did not change the mRNA levels of TNF-alpha, TGF-beta, and PDGF, as seen on Northern blots made with the lung RNA. Pulmonary platelet trapping, which was increased by a bleomycin injection, was decreased by an infusion of bombesin as demonstrated by the distribution of 111In-labeled platelets. This study indicates that bombesin act as an inhibitor of the development of pulmonary fibrosis, possibly by decreasing pulmonary platelet trapping.


Subject(s)
Bleomycin , Bombesin/pharmacology , Pulmonary Fibrosis/chemically induced , Animals , Antibodies, Monoclonal , Blood Platelets/drug effects , Bronchoalveolar Lavage Fluid/cytology , Cell Count , Cytokines/genetics , Hydroxyproline/metabolism , Immunohistochemistry , Lung/metabolism , Mice , Mice, Inbred C57BL , Mice, Inbred CBA , Pulmonary Fibrosis/mortality , RNA, Messenger/metabolism , Survival Analysis
SELECTION OF CITATIONS
SEARCH DETAIL
...