Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Int J Endocrinol ; 2019: 4687951, 2019.
Article in English | MEDLINE | ID: mdl-31662746

ABSTRACT

OBJECTIVE: To investigate the feasibility of near-infrared autofluorescence (AF) and indocyanine green (ICG) fluorescence to identify parathyroid glands intraoperatively. METHODS: Fluorescence imaging was carried out during open parathyroid and thyroid surgery. After visual identification, parathyroid glands were exposed to near-infrared (NIR) light with a wavelength between 690 and 770 nm. The camera of the Storz® NIR/ICG endoscopic system used detects NIR light as a blue signal. Therefore, parathyroid AF was expected to be displayed in the blue color channel in contrast to the surrounding tissue. Following AF imaging, a bolus of 5 mg ICG was applied intravenously. ICG fluorescence was detected using the same NIR/ICG imaging system. Well-vascularized parathyroid glands were expected to show a strong fluorescence in contrast to surrounding lymphatic and adipose tissue. RESULTS: We investigated 78 parathyroid glands from 50 patients. 64 parathyroid glands (82%) displayed AF showing the typical bluish violet color. 63 parathyroid glands (81%) showed a strong and persistent fluorescence after application of ICG. The sensitivity of identifying a parathyroid gland by AF was 82% (64 true positive and 14 false negative results), while ICG imaging showed a sensitivity of 81% (63 true positive and 15 false negative results). The Fisher exact test revealed no significant difference between both groups at p < 0.05. Neither lymph nodes nor adipose tissue revealed substantial AF or ICG fluorescence. CONCLUSION: AF and ICG fluorescence reveal a high degree of sensitivity in identifying parathyroid glands. Further, ICG imaging facilitates the assessment of parathyroid perfusion. However, in the current setting both techniques are not suitable as screening tools to identify parathyroid glands at an early stage of the operation.

2.
Blood ; 122(5): 770-80, 2013 Aug 01.
Article in English | MEDLINE | ID: mdl-23757732

ABSTRACT

In vitro studies suggest that leukocytes locomote in an ameboid fashion independently of pericellular proteolysis. Whether this motility pattern applies for leukocyte migration in inflamed tissue is still unknown. In vivo microscopy on the inflamed mouse cremaster muscle revealed that blockade of serine proteases or of matrix metalloproteinases (MMPs) significantly reduces intravascular accumulation and transmigration of neutrophils. Using a novel in vivo chemotaxis assay, perivenular microinjection of inflammatory mediators induced directional interstitial migration of neutrophils. Blockade of actin polymerization, but not of actomyosin contraction abolished neutrophil interstitial locomotion. Multiphoton laser scanning in vivo microscopy showed that the density of the interstitial collagen network increases in inflamed tissue, thereby providing physical guidance to infiltrating neutrophils. Although neutrophils locomote through the interstitium without pericellular collagen degradation, inhibition of MMPs, but not of serine proteases, diminished their polarization and interstitial locomotion. In this context, blockade of MMPs was found to modulate expression of adhesion/signaling molecules on neutrophils. Collectively, our data indicate that serine proteases are critical for neutrophil extravasation, whereas these enzymes are dispensable for neutrophil extravascular locomotion. By contrast, neutrophil interstitial migration strictly relies on actin polymerization and does not require the pericellular degradation of collagen fibers but is modulated by MMPs.


Subject(s)
Chemotaxis, Leukocyte/physiology , Inflammation/immunology , Matrix Metalloproteinases/physiology , Neutrophil Infiltration/physiology , Aminocaproates/pharmacology , Animals , Aprotinin/pharmacology , Chemotaxis, Leukocyte/drug effects , Immune System Diseases/metabolism , Immune System Diseases/pathology , Inflammation/metabolism , Leukocyte Disorders/metabolism , Leukocyte Disorders/pathology , Male , Matrix Metalloproteinases/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neutrophil Infiltration/drug effects , Neutrophil Infiltration/immunology , Peritonitis/immunology , Peritonitis/pathology , Tranexamic Acid/pharmacology , Transcellular Cell Migration/drug effects , Transcellular Cell Migration/immunology
3.
Circulation ; 124(17): 1848-59, 2011 Oct 25.
Article in English | MEDLINE | ID: mdl-21969013

ABSTRACT

BACKGROUND: Urokinase-type plasminogen activator (uPA) has recently been implicated in the pathogenesis of ischemia-reperfusion (I/R) injury. The underlying mechanisms remain largely unclear. METHODS AND RESULTS: Using in vivo microscopy on the mouse cremaster muscle, I/R-elicited firm adherence and transmigration of neutrophils were found to be significantly diminished in uPA-deficient mice and in mice treated with the uPA inhibitor WX-340, but not in uPA receptor (uPAR)-deficient mice. Interestingly, postischemic leukocyte responses were significantly reduced on blockade of the integrin CD11b/Mac-1, which also serves as uPAR receptor. Using a cell transfer technique, postischemic adherence and transmigration of wild-type leukocytes were significantly decreased in uPA-deficient animals, whereas uPA-deficient leukocytes exhibited a selectively reduced transmigration in wild-type animals. On I/R or stimulation with recombinant uPA, >90% of firmly adherent leukocytes colocalized with CD31-immunoreactive endothelial junctions as detected by in vivo fluorescence microscopy. In a model of hepatic I/R, treatment with WX-340 significantly attenuated postischemic neutrophil infiltration and tissue injury. CONCLUSIONS: Our data suggest that endothelial uPA promotes intravascular adherence, whereas leukocyte uPA facilitates the subsequent paracellular transmigration of neutrophils during I/R. This process is regulated via CD11b/Mac-1, and does not require uPAR. Pharmacological blockade of uPA interferes with these events and effectively attenuates postischemic tissue injury.


Subject(s)
Macrophage-1 Antigen/physiology , Neutrophils/cytology , Neutrophils/physiology , Receptors, Urokinase Plasminogen Activator/physiology , Transendothelial and Transepithelial Migration/physiology , Urokinase-Type Plasminogen Activator/physiology , Animals , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neutrophil Infiltration/physiology , Random Allocation
4.
PLoS One ; 6(2): e17229, 2011 Feb 22.
Article in English | MEDLINE | ID: mdl-21364954

ABSTRACT

Clinical trials revealed beneficial effects of the broad-spectrum serine protease inhibitor aprotinin on the prevention of ischemia-reperfusion (I/R) injury. The underlying mechanisms remained largely unclear. Using in vivo microscopy on the cremaster muscle of male C57BL/6 mice, aprotinin as well as inhibitors of the serine protease plasmin including tranexamic acid and ε-aminocaproic acid were found to significantly diminish I/R-elicited intravascular firm adherence and (subsequent) transmigration of neutrophils. Remodeling of collagen IV within the postischemic perivenular basement membrane was almost completely abrogated in animals treated with plasmin inhibitors or aprotinin. In separate experiments, incubation with plasmin did not directly activate neutrophils. Extravascular, but not intravascular administration of plasmin caused a dose-dependent increase in numbers of firmly adherent and transmigrated neutrophils. Blockade of mast cell activation as well as inhibition of leukotriene synthesis or antagonism of the platelet-activating-factor receptor significantly reduced plasmin-dependent neutrophil responses. In conclusion, our data suggest that extravasated plasmin(ogen) mediates neutrophil recruitment in vivo via activation of perivascular mast cells and secondary generation of lipid mediators. Aprotinin as well as the plasmin inhibitors tranexamic acid and ε-aminocaproic acid interfere with this inflammatory cascade and effectively prevent postischemic neutrophil responses as well as remodeling events within the vessel wall.


Subject(s)
Antifibrinolytic Agents/pharmacology , Leukocytes/drug effects , Microvessels/drug effects , Myocardial Reperfusion Injury/physiopathology , Neutrophil Infiltration/drug effects , Aminocaproic Acid/pharmacology , Animals , Aprotinin/pharmacology , Drug Evaluation, Preclinical , Leukocyte Count , Leukocytes/cytology , Leukocytes/physiology , Male , Mice , Mice, Inbred C57BL , Microvessels/immunology , Microvessels/pathology , Microvessels/physiopathology , Myocardial Reperfusion Injury/blood , Myocardial Reperfusion Injury/immunology , Myocardial Reperfusion Injury/rehabilitation , Serine Proteinase Inhibitors/pharmacology , Tranexamic Acid/pharmacology
5.
Biomaterials ; 31(26): 6692-700, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20619783

ABSTRACT

The increasing interest in biomedical applications of semiconductor quantum dots (QDs) is closely linked to the use of surface modifications to target specific sites of the body. The immense surface area of vascular endothelium is a possible interaction platform with systemically administered QDs. Therefore, the aim of this study was to investigate the microvascular distribution of neutral, cationic, and anionic QDs in vivo. QDs with carboxyl-, amine- and polyethylene glycol surface coatings were injected into the blood circulation of mice. In vivo microscopy of the cremaster muscle, two-photon microscopy of skeletal and heart muscle, as well as quantitative fluorescence measurements of blood, excreta, and tissue samples were performed. Transmission electron microscopy was used to detect QDs at the cellular level. The in vitro association of QDs with cultured endothelial cells was investigated by flow cytometry and confocal microscopy. Anionic QDs exhibited a very low residence time in the blood stream, preferably accumulated in organs with a prominent mononuclear phagocytic component, but were also found in other tissues with low phagocytic properties where they were predominantly associated with capillary endothelium. This deposition behavior was identified as a new, phagocyte-independent principle contributing to the rapid clearance of anionic QDs from the circulation.


Subject(s)
Endothelium, Vascular/metabolism , Quantum Dots , Animals , Anions , Endothelial Cells/cytology , Endothelial Cells/metabolism , Endothelial Cells/ultrastructure , Hemodynamics , Injections, Intra-Arterial , Kinetics , Male , Mice , Mice, Inbred C57BL , Microscopy , Microvessels/cytology , Microvessels/metabolism , Muscle, Skeletal/blood supply , Muscle, Skeletal/cytology , Muscle, Skeletal/metabolism , Myocardium/cytology , Myocardium/metabolism , Myocardium/ultrastructure , Photons , Tissue Distribution
6.
Toxicology ; 269(2-3): 148-54, 2010 Mar 10.
Article in English | MEDLINE | ID: mdl-19698757

ABSTRACT

OBJECTIVES: Although ambient nanoparticles have been shown to exert prothrombotic effects, manufactured nanoparticles are in this aspect less well investigated. Thus, the aim of this study was to characterize the effects of diesel, titanium dioxide rutile, and single-walled carbon nanotube nanoparticles on (i) platelet activation in vitro and (ii) on macro- and microcirculatory thrombus formation in vivo. METHODS: Platelet P-selectin expression was measured by flow cytometry after incubation of whole blood with diesel (0.1mg/mL), titanium dioxide (0.1mg/mL) or single-walled nanotubes (0.001-0.1mg/mL). Platelet-granulocyte complexes were analyzed in whole blood and platelet aggregometry was performed with platelet-rich plasma. Upon systemic administration of nanoparticles (1mg/kg) to anesthetized mice, ferric chloride-induced thrombus formation was measured in small mesenteric arteries using in vivo microscopy. In separate experiments, diesel (1mg/kg), titanium dioxide (1mg/kg), or single-walled nanotubes (0.01-1mg/kg) were injected into anesthetized mice and light/dye-induced thrombus formation was investigated in the cremasteric microcirculation. RESULTS: Diesel and titanium dioxide nanoparticles did not activate platelets or exert prothrombotic effects. In contrast, single-walled nanotubes significantly increased platelet P-selectin expression, the number of platelet-granulocyte complexes, and platelet aggregability in vitro, and reduced the occlusion time in mesenteric arteries as well as in cremasteric arterioles. CONCLUSION: Our study shows that single-walled carbon nanotubes, but not diesel or titanium dioxide nanoparticles, induce platelet activation in vitro and exert prothrombotic effects in the microcirculation in vivo.


Subject(s)
Blood Platelets/drug effects , Microcirculation/drug effects , Nanotubes, Carbon/toxicity , Platelet Activation/drug effects , Thrombosis/chemically induced , Animals , Blood Cell Count , Blood Platelets/metabolism , Granulocytes/metabolism , Male , Mice , Mice, Inbred C57BL , P-Selectin/metabolism , Titanium/toxicity , Vehicle Emissions/toxicity
7.
Arterioscler Thromb Vasc Biol ; 29(11): 1787-93, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19608967

ABSTRACT

OBJECTIVE: Although the chemokines monocyte chemoattractant protein-1 (Ccl2/JE/MCP-1) and macrophage inflammatory protein-1alpha (Ccl3/MIP-1alpha) have recently been implicated in neutrophil migration, the underlying mechanisms remain largely unclear. METHODS AND RESULTS: Stimulation of the mouse cremaster muscle with Ccl2/JE/MCP-1 or Ccl3/MIP-1alpha induced a significant increase in numbers of firmly adherent and transmigrated leukocytes (>70% neutrophils) as observed by in vivo microscopy. This increase was significantly attenuated in mice receiving an inhibitor of RNA transcription (actinomycin D) or antagonists of platelet activating factor (PAF; BN 52021) and leukotrienes (MK-886; AA-861). In contrast, leukocyte responses elicited by PAF and leukotriene-B(4) (LTB(4)) themselves were not affected by actinomycin D, BN 52021, MK-886, or AA-861. Conversely, PAF and LTB(4), but not Ccl2/JE/MCP-1 and Ccl3/MIP-1alpha, directly activated neutrophils as indicated by shedding of CD62L and marked upregulation of CD11b. Moreover, Ccl2/JE/MCP-1- and Ccl3/MIP-1alpha-elicited leakage of fluorescein isothiocyanate dextran as well as collagen IV remodeling within the venular basement membrane were completely absent in neutrophil-depleted mice. CONCLUSIONS: Ccl2/JE/MCP-1 and Ccl3/MIP-1alpha mediate firm adherence and (subsequent) transmigration of neutrophils via protein synthesis and secondary generation of leukotrienes and PAF, which in turn directly activate neutrophils. Thereby, neutrophils facilitate basement membrane remodeling and promote microvascular leakage.


Subject(s)
Chemokine CCL2/metabolism , Chemokine CCL3/metabolism , Lipoxygenase Inhibitors/pharmacology , Neutrophil Infiltration/physiology , Protein Biosynthesis/drug effects , 1-Alkyl-2-acetylglycerophosphocholine Esterase/pharmacology , Animals , Benzoquinones/pharmacology , Cells, Cultured , Chemokine CCL2/pharmacology , Chemokine CCL3/pharmacology , Chemotaxis, Leukocyte/drug effects , Chemotaxis, Leukocyte/physiology , Dactinomycin/pharmacology , Disease Models, Animal , Indoles/pharmacology , Indomethacin/pharmacology , Leukotriene B4/pharmacology , Male , Mice , Mice, Inbred BALB C , Neutrophil Infiltration/drug effects , Random Allocation , Sensitivity and Specificity
SELECTION OF CITATIONS
SEARCH DETAIL
...