Anaphylaxis-induced mesenteric vascular permeability, granulocyte adhesion, and platelet aggregates in rat

1998 ◽  
Vol 275 (1) ◽  
pp. H274-H284 ◽  
Author(s):  
Geoffrey D. Withers ◽  
Paul Kubes ◽  
Geoffrey Ibbotson ◽  
R. Brent Scott

This study investigates the response of small venules to IgE-dependent, antigen-mediated mast cell activation. Intravital microscopy was utilized to visualize 25- to 40-μm mesenteric venules, mast cell degranulation (on-line detection), vascular permeability changes (albumin leakage), leukocyte adhesion, and the formation of platelet aggregates in rats sensitized with 10 μg of intraperitoneal egg albumin (EA) in saline- or sham-sensitized (saline alone) rats. Sensitized rats challenged with EA (1 mg/ml superfusing mesentery), but not sensitized rats challenged with BSA or sham-sensitized rats challenged with EA, exhibited mast cell degranulation with significant time-dependent increases in vascular permeability (inhibited by diphenhydramine, salbutamol, and indomethacin), leukocyte adhesion (inhibited by Web-2086), and the formation of cellular aggregates (platelet), which were associated with intermittent obstruction of venular flow. Anti-platelet antibody, but not anti-neutrophil antibody or fucoidin (selectin antagonist), prevented platelet aggregate formation. Compound 48/80-induced mast cell degranulation caused similar changes in permeability (via different mediators) and leukocyte adhesion but did not induce platelet aggregation. EA-induced platelet aggregation was not inhibited by any of the mediators tested, and platelets isolated from sensitized rats failed to aggregate in response to direct EA challenge, suggesting release of an unidentified inflammatory mediator as the factor initiating platelet aggregation.

2003 ◽  
Vol 94 (1) ◽  
pp. 325-334 ◽  
Author(s):  
Dawn R. S. Steiner ◽  
Norberto C. Gonzalez ◽  
John G. Wood

Systemic hypoxia produces an inflammatory response characterized by increases in reactive O2 species (ROS), venular leukocyte-endothelial adherence and emigration, and vascular permeability. Inflammation is typically initiated by mediators released from activated perivascular cells that generate the chemotactic gradient responsible for extravascular leukocyte accumulation. These experiments were directed to study the possible participation of mast cells in hypoxia-induced microvascular inflammation. Mast cell degranulation, ROS levels, leukocyte adherence and emigration, and vascular permeability were studied in the mesenteric microcirculation by using intravital microscopy of anesthetized rats. The main findings were 1) activation of mast cells with compound 48/80 in normoxia produced microvascular effects similar, but not identical, to those of hypoxia; 2) systemic hypoxia resulted in rapid mast cell degranulation; 3) blockade of mast cell degranulation with cromolyn prevented or attenuated the hypoxia-induced increases in ROS, leukocyte adherence/emigration, and vascular permeability; and 4) mast cell degranulation during hypoxia was prevented by administration of the antioxidant lipoic acid and of nitric oxide. These results show that mast cells play a key role in hypoxia-induced inflammation and suggest that alterations in the ROS-nitric oxide balance may be involved in mast cell activation during hypoxia.


Circulation ◽  
2007 ◽  
Vol 116 (suppl_16) ◽  
Author(s):  
Ilze Bot ◽  
Saskia C de Jager ◽  
Alma Zernecke ◽  
Christian Weber ◽  
Theo J van Berkel ◽  
...  

Activated mast cells have been identified in the perivascular tissue of human coronary artery plaques. As mast cells have been described to release a whole array of chemokines including interleukin 8 (IL-8) and MIP1 α, we propose that activated mast cells play a pivotal role in leukocyte recruitment at advanced stages of atherosclerotic plaque development. Peritoneal mast cells of either C57Bl/6 or mast cell deficient Kit(W −sh /W −sh ) mice were activated by injection of compound 48/80 (1.2 mg/kg). Interestingly, mast cell activation led to a massive neutrophil influx into the peritoneal cavity at 3 hours after activation (controls: 1 ± 0.7*10 4 Gr1 + -neutrophils/ml up to 8 ± 0.2*10 4 Gr1 + neutrophils/ml at 3 hours after activation, *P<0.05), while neutrophil numbers in Kit(W −sh /W −sh ) mice were not affected by compound 48/80 administration. Moreover, increased levels of CXCR2 + Gr1 + neutrophils (t=0: 0.55 ± 0.07% versus t=3 hours: 1.00 ± 0.12%, *P<0.05) were observed after mast cell activation. Next, we investigated whether mast cell activation also translated in induced leukocyte adhesion to advanced atherosclerotic plaques. Adventitial mast cells of advanced collar aided carotid artery plaques were activated by local application of a dinitrophenyl-BSA (DNP) challenge in ApoE −/− mice. Three days later, the carotid artery segments carrying the plaques were isolated and perfused ex vivo with rhodamine labeled leukocytes, showing a dramatically increased number of adherent leukocytes after mast cell activation (49 ± 6 versus 19 ± 4 leukocytes/microscopic field for DNP versus control plaques, respectively, **P<0.001). Strikingly, antibody blockade of either the CXCR2 or VCAM-1 receptor VLA-4 on labeled leukocytes completely inhibited leukocyte adhesion to the atherosclerotic plaque (*P<0.05), while blockade of CCR1, -3 and -5 with Met-RANTES had no effect. In conclusion, our data suggest that chemokines such as IL-8 released from activated perivascular mast cells induce leukocyte recruitment and adhesion to the atherosclerotic plaque, aggravating the ongoing inflammatory response and thus effecting plaque destabilization. We propose that mast cell stabilization could be a new therapeutic approach in the prevention of acute coronary syndromes.


1993 ◽  
Vol 264 (5) ◽  
pp. H1346-H1354 ◽  
Author(s):  
B. R. Ito ◽  
R. L. Engler ◽  
U. del Balzo

Activated complement component C5a causes myocardial ischemia mediated by thromboxane (Tx) A2 and leukotrienes C4/D4. Blood cells are not involved in either the mediator release or the myocardial effects of C5a, suggesting that a C5a-sensitive, cardiac resident inflammatory cell is responsible. The goals of this study were to determine whether 1) cardiac mast cell activation accompanies the C5a response, 2) inhibition of mast cell degranulation inhibits the response, and 3) histamine release plays a role in the C5a-induced myocardial ischemia. The left anterior descending coronary artery (LAD) of open-chest pigs (n = 13) was perfused with arterial blood at constant pressure (95 mmHg). Coronary blood flow (CBF) was measured (in-line flowmeter) and regional function [percent segment shortening (%SS)] determined with sonomicrometry. A coronary vein was cannulated for measurement of plasma TxB2 and histamine (a marker of mast cell degranulation). Intracoronary C5a (500 ng) decreased coronary blood flow (45% of preinfusion levels) and LAD %SS (65% of preinfusion) and was accompanied by increases in coronary venous TxB2 (delta 63.3 ng/ml) and histamine (delta 200 nM). Mast cell inhibition with lodoxamide (2 mg/kg iv, n = 8) attenuated the C5a-induced fall in CBF (14 vs. 53% decrease, P < 0.01) and %SS (10 vs. 38% decrease, P < 0.01) and also reduced the C5a-induced increase in both coronary venous histamine (delta 26 vs. 278 nM, P < 0.05) and TxB2 (delta 0.34 vs. 63.3 ng/ml, P < 0.01). However, histamine H1 (pyrilamine) and H2 (ranitidine) receptor blockade had no effect on the C5a-induced fall in CBF or LAD %SS.(ABSTRACT TRUNCATED AT 250 WORDS)


1999 ◽  
Vol 86 (1) ◽  
pp. 202-210 ◽  
Author(s):  
N. Noviski ◽  
J. P. Brewer ◽  
W. A. Skornik ◽  
S. J. Galli ◽  
J. M. Drazen ◽  
...  

Exposure to ambient ozone (O3) is associated with increased exacerbations of asthma. We sought to determine whether mast cell degranulation is induced by in vivo exposure to O3in mice and whether mast cells play an essential role in the development of pulmonary pathophysiological alterations induced by O3. For this we exposed mast cell-deficient WBB6F1- kitW/ kitW-v( kitW/ kitW-v) mice and the congenic normal WBB6F1(+/+) mice to air or to 1 or 3 parts/million O3for 4 h and studied them at different intervals from 4 to 72 h later. We found evidence of O3-induced cutaneous, as well as bronchial, mast cell degranulation. Polymorphonuclear cell influx into the pulmonary parenchyma was observed after exposure to 1 part/milllion O3only in mice that possessed mast cells. Airway hyperresponsiveness to intravenous methacholine measured in vivo under pentobarbital anesthesia was observed in both kitW/ kitW-vand +/+ mice after exposure to O3. Thus, although mast cells are activated in vivo by O3and participate in O3-induced polymorphonuclear cell infiltration into the pulmonary parenchyma, they do not participate detectably in the development of O3-induced airway hyperresponsiveness in mice.


2014 ◽  
Vol 2014 ◽  
pp. 1-9 ◽  
Author(s):  
Constantin Căruntu ◽  
Daniel Boda ◽  
Sorin Musat ◽  
Ana Căruntu ◽  
Eugen Mandache

Mast cells play a key role in modulation of stress-induced cutaneous inflammation. In this study we investigate the impact of repeated exposure to stress on mast cell degranulation, in both hairy and glabrous skin. Adult male Wistar rats were randomly divided into four groups: Stress 1 day(n=8), Stress 10 days(n=7), Stress 21 days(n=6), and Control(n=8). Rats in the stress groups were subjected to 2 h/day restraint stress. Subsequently, glabrous and hairy skin samples from animals of all groups were collected to assess mast cell degranulation by histochemistry and transmission electron microscopy. The impact of stress on mast cell degranulation was different depending on the type of skin and duration of stress exposure. Short-term stress exposure induced an amplification of mast cell degranulation in hairy skin that was maintained after prolonged exposure to stress. In glabrous skin, even though acute stress exposure had a profound stimulating effect on mast cell degranulation, it diminished progressively with long-term exposure to stress. The results of our study reinforce the view that mast cells are active players in modulating skin responses to stress and contribute to further understanding of pathophysiological mechanisms involved in stress-induced initiation or exacerbation of cutaneous inflammatory processes.


Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 374-374
Author(s):  
Lucile Vincent ◽  
Julia Nguyen ◽  
Derek Vang ◽  
Oludare B Taiwo ◽  
Kathryn Luk ◽  
...  

Abstract Abstract 374 Sickle cell disease (SCD) is associated with inflammation, endothelial dysfunction and pain. We observed increased immunoreactivity (ir) of pro-inflammatory and vasoactive neuropeptides, substance P (SP) and calcitonin-gene related peptide (CGRP) accompanied by decreased mu opioid receptor (MOR)-ir in the skin of sickle as compared to control mice (Kohli et al., Blood 2010). SP activates mast cells (MC), which are tissue resident leukocytes, leading to the release of inflammatory cytokines, tryptase and neuropeptides. SP also stimulates vascular permeability resulting in plasma extravasation and neurogenic inflammation. We hypothesized that pain in SCD is associated with a persistent feed-forward cycle of mast cell degranulation and neurogenic inflammation characterized by increased release of SP and CGRP from activated nociceptors in the skin leading to neuroinflammation, plasma extravasation and pain. We examined this hypothesis using sickle (HbSS-BERK) and control (HbAA-BERK) mice expressing sickle and normal human hemoglobin, respectively; and MOR-knockout (MOR-KO) mice with their wild type 129S6 controls. We developed an ex-vivo system to analyze the release of inflammatory cytokines, mast cell degranulation markers (tryptase and beta-hexosaminidase) and neuropeptides in skin biopsies. Neurogenic inflammation was studied in vivo using the Miles' assay. Evans blue was injected into the tail vein and its extravasation in skin evoked by stimulation with SP and capsaicin was quantified. Skin biopsies from sickle mice exhibited constitutively enhanced release of several cytokines (IL6, MCP-1, TNFalpha, MIP-1alpha, GM-CSF, RANTES, etc), tryptase and the neuropeptides SP, and CGRP as compared to control mouse skin (p<0.05 for each). Increased RANTES and GM-CSF are suggestive of mast cell recruitment. Mast cell tryptase-ir was increased 2-fold while MOR-ir (but not delta- or kappa-OR-ir), was reduced by ∼50% in the skin of sickle as compared to control mice, suggestive of enhanced MC degranulation in sickle. In MC cultures prepared from sickle skin increased c-kit/CD117-, FCeR- and tryptase-ir were observed as compared to control mouse MCs. The plasma of sickle exhibited a ∼60–80% increase in MC degranulation markers, tryptase and beta-hexosaminidase, acute phase protein, serum amyloid protein, and neuropeptides, SP and CGRP, as compared to control mice (p<0.01 for each). These correlative molecular changes in the plasma and skin were accompanied by increased SP- and capsaicin-induced Evans blue dye leakage in the skin of sickle mice suggestive of neurogenic inflammation as compared to control (p<0.001 for each). MOR-KO mice also exhibited increased SP- and CGRP-ir in the skin and neurogenic inflammation, indicative of a contribution by MOR to the neuroinflamamtory process. In sickle mice treated with the mast cell stabilizer cromolyn sodium (CS), or the c-kit inhibitor, Imatinib, for 5 days, the inflammatory cytokine and neuropeptide release from the skin and the neurogenic inflammation were ameliorated as compared to vehicle (p<0.01). Additionally, morphine at a dose of 10 mg/Kg was ineffective in treating tonic cutaneous and thermal hyperalgesia, but effectively reduced hyperalgesia in CS and Imatinib treated sickle mice. Thus, MC degranulation contributes to neurogenic inflammation and pain in sickle mice. Imatinib treatment by itself reduced tonic hyperalgesia and significantly decreased GM-CSF release from the skin (p<0.05) correlative to the reduced white blood cell (WBC) count in sickle mice vs vehicle. In addition to inhibiting MC activity, Imatinib may be inhibiting protein tyrosine kinases involved in cytokine processing, vascular function and nociception. Together, our observations demonstrate that MCs contribute to a vicious cycle of pain and neurogenic inflammation mediated by increased neuropeptides in SCD. It is likely that mast cell inhibitors such as Imatinib may have a therapeutic effect on pain, inflammation and vascular dysfunction in SCD by reducing mast cell activation and neurogenic inflammation. Since, Imatinib decreased GM-CSF levels and WBC, it may even increase HbF levels, which are negatively regulated by GM-CSF in SCD. We therefore speculate that therapies targeting mast cells may potentiate therapeutic outcomes of analgesics, anti-inflammatory agents and Hydroxyurea® in SCD. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2012 ◽  
Vol 119 (14) ◽  
pp. 3306-3314 ◽  
Author(s):  
Jinwook Shin ◽  
Hongjie Pan ◽  
Xiao-Ping Zhong

Abstract Mast cells play critical roles in allergic disorders and asthma. The importance of tuberous sclerosis complex 1/2-mammalian target of rapamycin (TSC1/2-mTOR) signaling in mast cells is unknown. Here, we report that TSC1 is a critical regulator for mTOR signaling in mast cells downstream of FcεRI and c-Kit, and differentially controls mast cell degranulation and cytokine production. TSC1-deficiency results in impaired mast cell degranulation, but enhanced cytokine production in vitro and in vivo after FcεRI engagement. Furthermore, TSC1 is critical for mast cell survival through multiple pathways of apoptosis including the down-regulation of p53, miR-34a, reactive oxygen species, and the up-regulation of Bcl-2. Together, these findings reveal that TSC1 is a critical regulator of mast cell activation and survival, suggesting the manipulation of the TSC1/2-mTOR pathway as a therapeutic strategy for mast cell-mediated diseases.


Sign in / Sign up

Export Citation Format

Share Document