Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.790
Filter
Add more filters

Publication year range
1.
Microvasc Res ; 154: 104694, 2024 07.
Article in English | MEDLINE | ID: mdl-38723844

ABSTRACT

INTRODUCTION: Critical illness is associated with organ failure, in which endothelial hyperpermeability and tissue edema play a major role. The endothelial angiopoietin/Tie2 system, a regulator of endothelial permeability, is dysbalanced during critical illness. Elevated circulating angiopoietin-2 and decreased Tie2 receptor levels are reported, but it remains unclear whether they cause edema independent of other critical illness-associated alterations. Therefore, we have studied the effect of angiopoietin-2 administration and/or reduced Tie2 expression on microvascular leakage and edema under normal conditions. METHODS: Transgenic male mice with partial deletion of Tie2 (heterozygous exon 9 deletion, Tie2+/-) and wild-type controls (Tie2+/+) received 24 or 72 pg/g angiopoietin-2 or PBS as control (n = 12 per group) intravenously. Microvascular leakage and edema were determined by Evans blue dye (EBD) extravasation and wet-to-dry weight ratio, respectively, in lungs and kidneys. Expression of molecules related to endothelial angiopoietin/Tie2 signaling were determined by ELISA and RT-qPCR. RESULTS: In Tie2+/+ mice, angiopoietin-2 administration increased EBD extravasation (154 %, p < 0.05) and wet-to-dry weight ratio (133 %, p < 0.01) in lungs, but not in the kidney compared to PBS. Tie2+/- mice had higher pulmonary (143 %, p < 0.001), but not renal EBD extravasation, compared to wild-type control mice, whereas a more pronounced wet-to-dry weight ratio was observed in lungs (155 %, p < 0.0001), in contrast to a minor higher wet-to-dry weight ratio in kidneys (106 %, p < 0.05). Angiopoietin-2 administration to Tie2+/- mice did not further increase pulmonary EBD extravasation, pulmonary wet-to-dry weight ratio, or renal wet-to-dry weight ratio. Interestingly, angiopoietin-2 administration resulted in an increased renal EBD extravasation in Tie2+/- mice compared to Tie2+/- mice receiving PBS. Both angiopoietin-2 administration and partial deletion of Tie2 did not affect circulating angiopoietin-1, soluble Tie2, VEGF and NGAL as well as gene expression of angiopoietin-1, -2, Tie1, VE-PTP, ELF-1, Ets-1, KLF2, GATA3, MMP14, Runx1, VE-cadherin, VEGFα and NGAL, except for gene and protein expression of Tie2, which was decreased in Tie2+/- mice compared to Tie2+/+ mice. CONCLUSIONS: In mice, the microvasculature of the lungs is more vulnerable to angiopoietin-2 and partial deletion of Tie2 compared to those in the kidneys with respect to microvascular leakage and edema.


Subject(s)
Angiopoietin-2 , Capillary Permeability , Lung , Receptor, TIE-2 , Animals , Receptor, TIE-2/metabolism , Receptor, TIE-2/genetics , Angiopoietin-2/metabolism , Angiopoietin-2/genetics , Male , Lung/blood supply , Lung/metabolism , Lung/pathology , Kidney/blood supply , Kidney/metabolism , Signal Transduction , Mice, Knockout , Mice , Mice, Inbred C57BL , Pulmonary Edema/metabolism , Pulmonary Edema/genetics , Pulmonary Edema/pathology , Pulmonary Edema/chemically induced , Pulmonary Edema/physiopathology , Disease Models, Animal , Edema/metabolism , Mice, Transgenic , Ribonuclease, Pancreatic
2.
Br J Clin Pharmacol ; 90(7): 1751-1755, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38770584

ABSTRACT

To our knowledge, no prior study has analysed a possible association between acetazolamide and pulmonary oedema. The aim of this study was to use data from the EudraVigilance to detect a safety signal for acetazolamide-induced pulmonary oedema. We performed a disproportionality analysis (case-noncase method), calculating reporting odds ratios (RORs) up to 22 February 2024. Among 11 684 208 spontaneous cases of adverse reactions registered in EudraVigilance, 38 275 were pulmonary oedemas. Acetazolamide was involved in 31 cases. In more than half of those cases, the patients received a single dose of acetazolamide after undergoing cataract surgery: latency was 10-90 min. Remarkably, there were five cases of positive rechallenge and six cases resulted in death. The ROR for acetazolamide was 3.63 (95% CI 2.55-5.17). Disproportionality was also observed in VigiBase®: ROR 4.44 (95% CI 3.34-5.90). Our study confirms a signal that suggests a risk of serious pulmonary oedema associated with acetazolamide.


Subject(s)
Acetazolamide , Databases, Factual , Pulmonary Edema , Humans , Acetazolamide/adverse effects , Pulmonary Edema/chemically induced , Pulmonary Edema/epidemiology , Male , Female , Middle Aged , Aged , Databases, Factual/statistics & numerical data , Adverse Drug Reaction Reporting Systems/statistics & numerical data , Adult , Carbonic Anhydrase Inhibitors/adverse effects , Carbonic Anhydrase Inhibitors/administration & dosage , Pharmacovigilance , Aged, 80 and over
3.
J Obstet Gynaecol Res ; 50(7): 1111-1117, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38627884

ABSTRACT

AIM: In Japan, unlike Western countries, tocolytic agents are administered in long-term protocols to treat threatened preterm labor. Evaluating the side effects of this practice is crucial. We examined whether ritodrine hydrochloride had been administered in cases of maternal death, aiming to investigate any relationship between ritodrine administration and maternal death. METHODS: This retrospective cohort study used reports of maternal deaths from multiple institutions in Japan between 2010 and 2020. Data on the reported cases were retrospectively analyzed, and data on the route of administration, administered dose, and clinical findings, including causes of maternal death, were extracted. The amount of tocolytic agents was compared between maternal deaths with ritodrine administration and those without. RESULTS: A total of 390 maternal deaths were reported to the Maternal Death Exploratory Committee in Japan during the study period. Ritodrine hydrochloride was administered in 32 of these cases. The frequencies (n) and median doses (range) of oral or intravenous ritodrine hydrochloride were 34.4% (11) and 945 (5-2100) mg and 84.4% (27) and 4032 (50-18 680) mg, respectively. Frequencies of perinatal cardiomyopathy, cerebral hemorrhage, diabetic ketoacidosis, and pulmonary edema as causes of maternal death were significantly higher with ritodrine administration than without it. CONCLUSIONS: Our results suggest a relationship between long-term administration of ritodrine hydrochloride and an increased risk of maternal death due to perinatal cardiomyopathy, cerebral hemorrhage, diabetic ketoacidosis, and pulmonary edema. In cases where ritodrine should be administered to prevent preterm labor, careful management and monitoring of maternal symptoms are required.


Subject(s)
Maternal Mortality , Ritodrine , Tocolytic Agents , Humans , Ritodrine/administration & dosage , Ritodrine/adverse effects , Tocolytic Agents/administration & dosage , Tocolytic Agents/adverse effects , Female , Pregnancy , Japan/epidemiology , Retrospective Studies , Adult , Obstetric Labor, Premature/drug therapy , Pulmonary Edema/mortality , Pulmonary Edema/chemically induced
4.
Ophthalmic Plast Reconstr Surg ; 40(2): e35-e36, 2024.
Article in English | MEDLINE | ID: mdl-37721321

ABSTRACT

A 72-year-old female with epiphora presented for outpatient punctoplasty with probing and lacrimal stent placement. Oxymetazoline was administered intranasally and the case was completed in standard fashion. Postoperatively, the patient desaturated with a workup revealing elevated cardiac enzymes, pulmonary congestion, and sinus bradycardia. However, the final cardiac testing was noncontributory, suggesting flash pulmonary edema secondary to intranasal oxymetazoline. This case highlights a rare presentation of pulmonary compromise secondary to oxymetazoline, emphasizing the importance of intraoperative and postoperative vigilance in simple outpatient procedures.


Subject(s)
Lacrimal Apparatus Diseases , Pulmonary Edema , Female , Humans , Aged , Oxymetazoline/adverse effects , Pulmonary Edema/chemically induced , Pulmonary Edema/diagnosis , Administration, Intranasal , Nose
5.
BMC Med ; 21(1): 47, 2023 02 10.
Article in English | MEDLINE | ID: mdl-36765407

ABSTRACT

BACKGROUND: Impaired respiratory function remains underrecognized in patients with type 2 diabetes (T2D), despite common pulmonary impairment. Meanwhile, there is little data available on the respiratory effects of sodium glucose cotransporter 2 inhibitors (SGLT2i). Hence, we examined the association between SGLT2i use and the risk of adverse respiratory events in a real-world setting. METHODS: We conducted a population-based, nationwide cohort study using an active-comparator new-user design and nationwide claims data of South Korea from January 2015 to December 2020. Among individuals aged 18 years or older, propensity score matching was done to match each new user of SGLT2is with dipeptidyl peptidase 4 inhibitors (DPP4is), with patients followed up according to an as-treated definition. The primary outcome was respiratory events, a composite endpoint of acute pulmonary edema, acute respiratory distress syndrome (ARDS), pneumonia, and respiratory failure. Secondary outcomes were the individual components of the primary outcome and in-hospital death. Cox models were used to estimate hazard ratios (HRs) and 95% CIs. RESULTS: Of 205,534 patient pairs in the propensity score matched cohort, the mean age of the entire cohort was 53.8 years and 59% were men, with a median follow-up of 0.66 years; all baseline covariates achieved balance between the two groups. Incidence rates for overall respiratory events were 4.54 and 7.54 per 1000 person-years among SGLT2i and DPP4i users, respectively, corresponding to a rate difference of 3 less events per 1000 person-years (95% CI - 3.44 to - 2.55). HRs (95% CIs) were 0.60 (0.55 to 0.64) for the composite respiratory endpoint, 0.35 (0.23 to 0.55) for acute pulmonary edema, 0.44 (0.18 to 1.05) for ARDS, 0.61 (0.56 to 0.66) for pneumonia, 0.49 (0.31 to 0.76) for respiratory failure, and 0.46 (0.41 to 0.51) for in-hospital death. Similar trends were found across individual SGLT2is, subgroup analyses of age, sex, history of comorbidities, and a range of sensitivity analyses. CONCLUSIONS: These findings suggest a lower risk of adverse respiratory events associated with patients with T2D initiating SGLT2is versus DPP4is. This real-world evidence helps inform patients, clinicians, and guideline writers regarding the respiratory effects of SGLT2i in routine practice.


Subject(s)
Diabetes Mellitus, Type 2 , Dipeptidyl-Peptidase IV Inhibitors , Pulmonary Edema , Respiratory Insufficiency , Sodium-Glucose Transporter 2 Inhibitors , Male , Humans , Middle Aged , Female , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/epidemiology , Dipeptidyl-Peptidase IV Inhibitors/adverse effects , Cohort Studies , Pulmonary Edema/chemically induced , Pulmonary Edema/complications , Hospital Mortality , Sodium-Glucose Transporter 2 Inhibitors/adverse effects , Respiratory Insufficiency/chemically induced , Respiratory Insufficiency/complications , Glucose , Sodium , Hypoglycemic Agents , Retrospective Studies
6.
Drug Chem Toxicol ; 46(5): 1024-1034, 2023 Nov.
Article in English | MEDLINE | ID: mdl-36069203

ABSTRACT

We aimed to observe the possible effects of melatonin (MLT) deprivation (pinealectomy) and exogenous MLT administration on pulmonary edema induced by alpha-naphthylthiourea (ANTU), a toxic chemical agent, in rats. Seventy animals were assigned to seven groups: control, sham pinealectomy (PINX), PINX, ANTU (10 mg/kg intraperitoneal on day 30), ANTU + MLT (10 mg/kg/day i.p. for 30 days), ANTU + PINX, and ANTU + PINX + MLT.In this study, pleural effusion (PE) formation, lung weight/body weight (LW/BW) and PE/BW ratios (fluid accumulation and weight values in the lungs) increase detected. Pre-ANTU MLT administration led to significant decreases in PE, LW/BW, and PE/BW levels. The inhibited glutathione (GSH) and superoxide dismutase (SOD) levels and high malondialdehyde (MDA) levels that ANTU increase lipid peroxidation in the study. MLT administration eliminated oxidative stress by reducing MDA and ameliorating GSH and SOD levels.Pre-ANTU MLT administration led to a significant decrease in interleukin-1 beta (IL-1ß) and tumor necrosis factor-alpha (TNF-α) levels in the lung when compared to the ANTU group without MLT administration. Post-pinealectomy ANTU administration significantly increased IL-1ß and TNF-α levels when compared to ANTU and MLT administration without pinealectomy. Diffused inflammatory cell infiltration, interstitial pulmonary edema, and histopathological congestion were observed after the administration of ANTU. Severity of the damage was elevated in the ANTU + PINX group. MLT treatment regressed pulmonary effusion and edema and improves lung structure. In brief, the findings suggested that MLT inhibited proinflammatory mediators and could serve as a therapeutic agent to prevent inflammatory disorders.


Subject(s)
Melatonin , Pulmonary Edema , Rats , Animals , Pulmonary Edema/chemically induced , Pulmonary Edema/prevention & control , Pulmonary Edema/pathology , Melatonin/pharmacology , Pinealectomy , Tumor Necrosis Factor-alpha , Thiourea/toxicity
7.
J Biochem Mol Toxicol ; 36(10): e23163, 2022 Oct.
Article in English | MEDLINE | ID: mdl-35844137

ABSTRACT

The renin-angiotensin system (RAS) is an important regulator in pulmonary physiology. In our study, we identified the efficacy of melatonin to control the RAS in cadmium (Cd) induced chronic lung injury in a mouse model. Swiss albino mice exposed to CdCl2 intraperitoneally (I.P.) (1 mg/kg b.w.; 12 weeks) showed increased release of lactate dehydrogenase in bronchoalveolar lavage fluid, generating reactive oxygen species, impaired antioxidant enzymes function, and disrupted alveolar structure along with increased expression of Angiotensin-II (Ang-II) in lung tissue. Cd-induced angiotensin-converting enzyme-2-Ang-II axis imbalance triggered the onset of Ang-II induced tumour necrosis factor alpha  (TNF-α) mediated necroptosis by upregulating the signalling molecules RIP-1, RIP-3, and p-mixed lineage kinase domain-like. In an in vitro study, colocalization of Ang-II-RIP-3 molecule in Cd intoxicated L-132 cells (human alveolar epithelial cell line), as well as pretreatment of Cd exposed cells with the inhibitor's captopril (10 µM), necrostatin-1 (50 µM), and etanercept (5 µg/ml) indicated TNF-α induced necroptotic cell death via activation of the key molecule, Ang-II. Moreover, Ang-II disrupted the alveolar-capillary barrier by decreasing tight junctional proteins (zonula occludens-1 and occludin) and endothelial VE-cadherin expression. The use of human umbilical vein endothelial cells as a model of junctional protein-expressing cells showed that captopril pretreatment (25 µM) restored VE-cadherin expression in Cd-treated human umbilical vein endothelial cells. In CdCl2 intoxicated mice, melatonin pretreatment (10 mg/kg b.w.; 12 weeks, I.P.) inhibited inflammatory mediators (TNF-α, interleukin [IL]-1ß, and IL-6) release and effectively suppressed (Cd-induced) Ang-II mediated necroptotic cell death and alveolar-capillary breaching due to Cd toxicity.


Subject(s)
Lung Injury , Melatonin , Pulmonary Edema , Angiotensin II/pharmacology , Animals , Antioxidants , Cadmium/toxicity , Captopril/pharmacology , Endothelial Cells/metabolism , Etanercept , Humans , Interleukin-6/metabolism , Lactate Dehydrogenases , Lung Injury/chemically induced , Lung Injury/drug therapy , Mice , Necroptosis , Occludin , Pulmonary Edema/chemically induced , Pulmonary Edema/drug therapy , Reactive Oxygen Species/metabolism , Tumor Necrosis Factor-alpha/metabolism
8.
BMC Pregnancy Childbirth ; 22(1): 636, 2022 Aug 12.
Article in English | MEDLINE | ID: mdl-35962336

ABSTRACT

BACKGROUND: A few studies have reported that maternal administration of antenatal corticosteroids increased the risk of pulmonary edema (PE). However, despite the increasing usage rate of betamethasone as antenatal corticosteroid, maternal administration of betamethasone as a risk factor for PE has not been well studied. This study aimed to evaluate how maternal backgrounds and complications, tocolytic agents, and betamethasone affect the incidence of PE during the perinatal period and determine the risk factor for PE. METHODS: This was a single-center retrospective cohort study in Kurashiki, Japan. The study subjects were patients who had been admitted to our hospital for perinatal management including pregnancy, delivery and puerperium between 2017 and 2020. The primary outcome measure was defined as the incidence of PE during hospitalization. First, in all study subjects, Cox proportional hazards model was used to determine the risk factor for PE during the perinatal period. Next, using propensity score matching, we divided the patients into the betamethasone and betamethasone-free groups and examined the association between betamethasone use and the incidence of PE with Cox proportional hazards model. RESULTS: During the study period, 4919 cases were hospitalized, and there were 16 PE cases (0.3%). In all analyzed subjects, the occurrence of PE was significantly associated with preeclampsia (hazard ratio 16.8, 95% confidence intervals (CI) 5.39-52.7, P < 0.001) and the combined use of the tocolytic agents such as ritodrine hydrochloride and magnesium sulfate, and betamethasone (hazard ratio 11.3, 95% CI 2.66-48.1, P = 0.001). In contrast, after propensity score matching, no statistically significant difference was found between the betamethasone and betamethasone-free groups in the incidence of PE (hazard ratio 3.19, 95% CI 0.67-15.3, P = 0.145). CONCLUSIONS: A combined use of tocolytic agents and antenatal corticosteroids such as betamethasone may be an independent risk factor for PE during the perinatal period. On the other hand, betamethasone use alone may not be associated with the incidence of PE. When tocolytic agents and betamethasone are administrated to pregnant women, it is important to pay attention to the appearance of maternal respiratory symptoms.


Subject(s)
Premature Birth , Pulmonary Edema , Tocolytic Agents , Adrenal Cortex Hormones/adverse effects , Betamethasone/adverse effects , Female , Humans , Japan/epidemiology , Pregnancy , Premature Birth/chemically induced , Pulmonary Edema/chemically induced , Pulmonary Edema/epidemiology , Retrospective Studies , Risk Factors , Tocolytic Agents/adverse effects
9.
Am J Emerg Med ; 53: 41-43, 2022 Mar.
Article in English | MEDLINE | ID: mdl-34973491

ABSTRACT

OBJECTIVE: Describe a series of patients who developed naloxone-associated pulmonary edema after recreational opioid use. DESIGN: Single center retrospective case series of patients who developed pulmonary edema following the prehospital administration of naloxone. SETTING: Academic, urban safety-net hospital. PATIENTS: Adults with recreational opioid overdose who developed naloxone-associated pulmonary edema, defined as the acute onset of respiratory distress, hypoxemia, and radiographic pulmonary edema after naloxone administration for opioid intoxication, provided that gas exchange and chest imaging rapidly improved and pulmonary aspiration of gastric contents was not clinically suspected. MEASUREMENTS AND MAIN RESULTS: Ten adults (median age 23 years, 90% male) met our case definition for naloxone-associated pulmonary edema. Implicated opioids were heroin in 8 patients and methadone and oxycodone in 1 patient each. The median total dose of naloxone was 4.25 mg (interquartile range [IQR] 3.3-9.8) prior to the onset of clinically-apparent pulmonary edema. Seven patients received invasive mechanical ventilation for a median of two days (IQR 0.8-5), one of whom received veno-venous extracorporeal membrane oxygenation support, and all survived to hospital discharge. CONCLUSIONS: Severe acute pulmonary edema may follow naloxone administration after recreational opioid overdose. Acute care clinicians should be aware of this potentially life-threatening adverse effect of naloxone.


Subject(s)
Drug Overdose , Opiate Overdose , Pulmonary Edema , Adult , Analgesics, Opioid/adverse effects , Drug Overdose/drug therapy , Female , Humans , Male , Naloxone/therapeutic use , Narcotic Antagonists/therapeutic use , Pulmonary Edema/chemically induced , Pulmonary Edema/drug therapy , Retrospective Studies , Young Adult
10.
BMC Pulm Med ; 22(1): 118, 2022 Mar 31.
Article in English | MEDLINE | ID: mdl-35361181

ABSTRACT

BACKGROUND: Noncardiogenic pulmonary edema (NCPE) is a rare and life-threatening allergy-like reaction to the intravascular injection of a nonionic radiographic agent. We first describe a very rare case of fatal NCPE after the intravenous injection of nonionic, iso-osmolar iodine contrast media. Case presentation A 55-year-old male patient was admitted to the hospital with esophageal cancer. After the intravenous administration of 100 mL iodixanol, the patient first exhibited digestive tract symptoms, including abdominal pain, diarrhea, and vomiting, with no dyspnea, rash, itching, or throat edema. He received anti-allergy treatment, but his symptoms did not improve; instead, he further developed pulmonary edema. Arterial blood gas analysis results were as follows: pH, 7.08; PO2, 70 mm Hg; PCO2, 40 mm Hg; and SaO2, 52%. Then, the patient received emergent tracheal intubation and ventilation to assist breathing, and he was transferred to the intensive care unit (ICU) for further treatment. In the ICU, the patient developed shock and respiratory and circulatory failure; therefore, he received shock resuscitation, acidosis correction, muscle relaxants to lower the work of breathing, and cardiotonic therapy. The patient eventually died. During the ICU period, emergency bedside color ultrasound showed a diffuse B line in both lungs, and the size of the cardiac cavity was normal, but the ventricular rate was extremely fast. Chest radiography showed pulmonary edema with a normal cardiac silhouette, and the brain natriuretic peptide (BNP) level was in the normal range. CONCLUSIONS: NCPE is a rare and critical allergy-like reaction to the use of a nonionic iso-osmolar radiocontrast contrast medium. Clinicians should pay very close attention to digestive tract manifestations during the medical observation of patients, as gastrointestinal manifestations may be the prodromal symptoms of NCPE caused by iso-osmolar contrast medium injection.


Subject(s)
Iodine , Pulmonary Edema , Contrast Media/adverse effects , Humans , Injections, Intravenous , Lung , Male , Middle Aged , Pulmonary Edema/chemically induced , Pulmonary Edema/diagnostic imaging
11.
An Acad Bras Cienc ; 94(suppl 4): e20211327, 2022.
Article in English | MEDLINE | ID: mdl-36449861

ABSTRACT

Acute lung injury is an inflammation that triggers acute respiratory distress syndrome with perialveolar neutrophil infiltration, alveolar-capillary barrier damage, and lung edema. Activation of the toll-like receptor 4 complex (TLR4/MD2) and its downstream signaling pathways are responsible for the cytokine storm and cause alveolar damage. Due to the complexity of this pulmonary inflammation, a defined pharmacotherapy has not been established. Thus, this study evaluated the anti-inflammatory potential of milonine, an alkaloid of Cissampelos sympodialis Eichl, in an experimental model of lung inflammation. BALB/c mice were lipopolysaccharide-challenged and treated with milonine at 2.0 mg/kg. Twenty-four hours later, the bronchoalveolar fluid, peripheral blood, and lungs were collected for cellular and molecular analysis. The milonine treatment decreased the cell migration (mainly neutrophils) to the alveoli, the pulmonary edema, and the cytokine levels (IL-1ß, IL-6, TNF-α). The systemic IL-6 level was also reduced. The milonine docking analysis demonstrated hydrophobic interaction at TLR4/MD2 groove with Ile124 and Phe126 amino acids. Indeed, the alkaloid downregulated the kinase-Akt and NF-κB through TLR4/MD2. Therefore, milonine is an effective inflammatory modulator being a potential molecule for the treatment of lung inflammation.


Subject(s)
Acute Lung Injury , Pulmonary Edema , Mice , Animals , NF-kappa B , Lipopolysaccharides , Toll-Like Receptor 4 , Proto-Oncogene Proteins c-akt , Interleukin-6 , Acute Lung Injury/chemically induced , Acute Lung Injury/drug therapy , Pulmonary Edema/chemically induced , Pulmonary Edema/drug therapy , Signal Transduction
12.
Bull Exp Biol Med ; 173(5): 623-627, 2022 Sep.
Article in English | MEDLINE | ID: mdl-36210422

ABSTRACT

We studied the content of aquaporin-5 (AQP5) and epithelial sodium channel (ENaC) in rat lungs during the development of toxic pulmonary edema (TPE) caused by intoxication with phosgene and perfluoroisobutylene (1.5 LC50). The lung body weight index (LBI) was calculated and histological examination of the lung tissues was performed. Localization and expression of AQP5 and ENaC were determined by immunohistochemistry. Intoxication led to a significant (p<0.05) increase in LBI and histological changes typical of TPE 1 and 3 h after the exposure. In 1 and 3 h after phosgene intoxication, the AQP5 and ENaC content significantly (p<0.05) increased in comparison with the control. Similar changes in the AQP5 and ENaC content were observed 1 and 3 h after exposure to perfluoroisobutylene. It was hypothesized that AQP5 plays an important role in the formation of TPE caused by intoxication with acylating pulmonotoxicants. An increase in the content of ENaC can be considered as a compensatory reaction of the body aimed at clearance of the alveolar fluid.


Subject(s)
Aquaporin 5 , Epithelial Sodium Channels , Fluorocarbons , Phosgene , Pulmonary Edema , Animals , Aquaporin 5/metabolism , Epithelial Sodium Channels/metabolism , Fluorocarbons/toxicity , Lung/metabolism , Phosgene/toxicity , Pulmonary Alveoli/metabolism , Pulmonary Edema/chemically induced , Pulmonary Edema/pathology , Rats
13.
Pflugers Arch ; 473(11): 1723-1735, 2021 11.
Article in English | MEDLINE | ID: mdl-34510286

ABSTRACT

Acute normobaric hypoxia may induce pulmonary injury with edema (PE) and inflammation. Hypoxia is accompanied by sympathetic activation. As both acute hypoxia and high plasma catecholamine levels may elicit PE, we had originally expected that adrenergic blockade may attenuate the severity of hypoxic pulmonary injury. In particular, we investigated whether administration of drugs with reduced fluid load would be beneficial with respect to both cardiocirculatory and pulmonary functions in acute hypoxia. Rats were exposed to normobaric hypoxia (10% O2) over 1.5 or 6 h and received 0.9% NaCl or adrenergic blockers either as infusion (1 ml/h, increased fluid load) or injection (0.5 ml, reduced fluid load). Control animals were kept in normoxia and received infusions or injections of 0.9% NaCl. After 6 h of hypoxia, LV inotropic function was maintained with NaCl injection but decreased significantly with NaCl infusion. Adrenergic blockade induced a similar LV depression when fluid load was low, but did not further deteriorate LV depression after 6 h of infusion. Reduced fluid load also attenuated pulmonary injury after 6 h of hypoxia. This might be due to an effective fluid drainage into the pleural space. Adrenergic blockade could not prevent PE. In general, increased fluid load and impaired LV inotropic function promote the development of PE in acute hypoxia. The main physiologic conclusion from this study is that fluid reduction under hypoxic conditions has a protective effect on cardiopulmonary function. Consequently, appropriate fluid management has particular importance to subjects in hypoxic conditions.


Subject(s)
Adrenergic Antagonists/pharmacology , Heart Ventricles/drug effects , Hypoxia/chemically induced , Pulmonary Edema/chemically induced , Animals , Female , Heart Ventricles/physiopathology , Hypoxia/physiopathology , Lung/drug effects , Lung/physiopathology , Pulmonary Edema/physiopathology , Rats , Rats, Sprague-Dawley
14.
Toxicol Appl Pharmacol ; 415: 115430, 2021 03 15.
Article in English | MEDLINE | ID: mdl-33524446

ABSTRACT

Air pollutants may increase risk for cardiopulmonary disease, particularly in susceptible populations with metabolic stressors such as diabetes and unhealthy diet. We investigated effects of inhaled ozone exposure and high-cholesterol diet (HCD) in healthy Wistar and Wistar-derived Goto-Kakizaki (GK) rats, a non-obese model of type 2 diabetes. Male rats (4-week old) were fed normal diet (ND) or HCD for 12 weeks and then exposed to filtered air or 1.0 ppm ozone (6 h/day) for 1 or 2 days. We examined pulmonary, vascular, hematology, and inflammatory responses after each exposure plus an 18-h recovery period. In both strains, ozone induced acute bronchiolar epithelial necrosis and inflammation on histopathology and pulmonary protein leakage and neutrophilia; the protein leakage was more rapid and persistent in GK compared to Wistar rats. Ozone also decreased lymphocytes after day 1 in both strains consuming ND (~50%), while HCD increased circulating leukocytes. Ozone increased plasma thrombin/antithrombin complexes and platelet disaggregation in Wistar rats on HCD and exacerbated diet effects on serum IFN-γ, IL-6, KC-GRO, IL-13, and TNF-α, which were higher with HCD (Wistar>GK). Ex vivo aortic contractility to phenylephrine was lower in GK versus Wistar rats at baseline(~30%); ozone enhanced this effect in Wistar rats on ND. GK rats on HCD had higher aortic e-NOS and tPA expression compared to Wistar rats. Ozone increased e-NOS in GK rats on ND (~3-fold) and Wistar rats on HCD (~2-fold). These findings demonstrate ways in which underlying diabetes and HCD may exacerbate pulmonary, systemic, and vascular effects of inhaled pollutants.


Subject(s)
Air Pollutants/toxicity , Aorta, Thoracic/drug effects , Cholesterol, Dietary/toxicity , Diabetes Mellitus, Type 2/complications , Diet, Atherogenic/adverse effects , Lung Injury/chemically induced , Lung/drug effects , Ozone/toxicity , Vascular Diseases/chemically induced , Animals , Aorta, Thoracic/metabolism , Aorta, Thoracic/physiopathology , Biomarkers/blood , Blood Platelets/drug effects , Blood Platelets/metabolism , Cholesterol, Dietary/metabolism , Cytokines/blood , Diabetes Mellitus, Type 2/blood , Disease Models, Animal , Inflammation Mediators/blood , Inhalation Exposure , Lung/metabolism , Lung/pathology , Lung Injury/blood , Lung Injury/pathology , Male , Necrosis , Pulmonary Edema/blood , Pulmonary Edema/chemically induced , Pulmonary Edema/pathology , Rats, Wistar , Vascular Diseases/blood , Vascular Diseases/physiopathology , Vasoconstriction/drug effects
15.
Toxicol Appl Pharmacol ; 422: 115460, 2021 07 01.
Article in English | MEDLINE | ID: mdl-33774062

ABSTRACT

To explore the protective mechanism of simvastatin in acute lung injury (ALI), the lipopolysaccharide (LPS) induced (5 mg/kg) ALI rat model was used to examine the effects of simvastatin. Following simvastatin treatment, the histopathological evaluation of lung tissues was made using hematoxylin and eosin (H&E) staining. Also, myeloperoxidase (MPO) activity and the levels of tumor necrosis factor-alpha (TNF-α), interleukin-1 beta (IL-1ß), and IL-10 were determined by ELISA. Blood gas analyses of arterial blood samples were performed to assess the pulmonary gas exchange. Moreover, the neutrophil count and total protein content were determined in the bronchoalveolar lavage (BAL) fluid. The ratio of wet lung to dry lung (W/D) and the alveolar fluid clearance (AFC) were calculated to estimate the severity of edema. Lastly, the levels of A2BAR, CFTR, claudin4, and claudin18 were also measured by qRT-PCR and Western blotting. Simvastatin treatment, in a dose-related manner, markedly improved the lung histological injury and decreased the levels of TNF-α, IL-1ß, and increased IL-10 in LPS induced ALI. Also, pulmonary neutrophil count was alleviated. Besides, a decreased ratio of W/D lung also confirmed the simvastatin intervention. Notably, simvastatin reduced the levels of A2BAR, CFTR, and claudin18 but upregulated claudin4 in lung tissues. Additionally, treatment with PSB1115, an antagonist of A2BAR, countered the protective effect of simvastatin in ALI. Our study demonstrates that simvastatin has a protective effect against LPS-induced ALI by activating A2BAR and should be exploited as a novel therapeutic target for the treatment of ALI.


Subject(s)
Acute Lung Injury/prevention & control , Adenosine A2 Receptor Agonists/pharmacology , Lung/drug effects , Receptor, Adenosine A2B/drug effects , Simvastatin/pharmacology , Acute Lung Injury/chemically induced , Acute Lung Injury/metabolism , Acute Lung Injury/pathology , Animals , Claudin-4/metabolism , Claudins/metabolism , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Cytokines/metabolism , Disease Models, Animal , Inflammation Mediators/metabolism , Lipopolysaccharides , Lung/metabolism , Lung/pathology , Male , Neutrophil Infiltration/drug effects , Pulmonary Edema/chemically induced , Pulmonary Edema/metabolism , Pulmonary Edema/pathology , Pulmonary Edema/prevention & control , Rats, Sprague-Dawley , Receptor, Adenosine A2B/metabolism , Signal Transduction
16.
BMC Neurol ; 21(1): 346, 2021 Sep 09.
Article in English | MEDLINE | ID: mdl-34503474

ABSTRACT

BACKGROUND: Recombinant tissue plasminogen activator (rt-pa) is the first-line drug for the treatment of acute ischemic stroke, and can lead to some complications.There were rare reports of death due to acute pulmonary edema during rt-pa thrombolysis treatment. CASE PRESENTATION: This study reports a 30-year-old man was diagnosed with acute ischemic stroke and underwent rt-pa thrombolytic therapy. Finally he died despite active rescue. CONCLUSIONS: The autopsy revealed that he died of acute pulmonary edema. This case suggests that it is necessary to pay close attention to the changes of vital signs during thrombolysis and be aware of possibility of pulmonary edema during thrombolysis.


Subject(s)
Brain Ischemia , Pulmonary Edema , Stroke , Adult , Brain Ischemia/drug therapy , Fibrinolytic Agents/adverse effects , Humans , Male , Pulmonary Edema/chemically induced , Pulmonary Edema/drug therapy , Recombinant Proteins/therapeutic use , Stroke/complications , Stroke/drug therapy , Thrombolytic Therapy/adverse effects , Tissue Plasminogen Activator/adverse effects , Treatment Outcome
17.
Am J Emerg Med ; 39: 256.e5-256.e8, 2021 01.
Article in English | MEDLINE | ID: mdl-33069545

ABSTRACT

Guanfacine is a central alpha-2 agonist often prescribed for Attention-deficit hyperactive disorder as well as tic disorder, with a usual dose of 1-4 mg per day. Due to its sympatholytic mechanism of action, Guanfacine can cause autonomic instability and hypotension. It can additionally cause cardiac dysfunction to include symptomatic bradycardias and contractility suppression. The authors present a case of a 17 year-old male with an ingestion of 80 mg of extended release Guanfacine with delayed onset cardiogenic pulmonary edema requiring mechanical ventilation. Previous pediatric ingestions have generated bradycardia, hypotension, and decreased level of consciousness, responsive to intravenous fluids, vasopressors, and occasionally naloxone. However, cardiogenic pulmonary edema from reduced cardiac contractility is a novel consequence of extended release Guanfacine ingestion. With Guanfacine's extended half-life, this unique case underscores the importance of emergency providers' familiarity with this toxidrome as well the necessity for prolonged, close observation following Guanfacine ingestion.


Subject(s)
Adrenergic alpha-2 Receptor Agonists/poisoning , Drug Overdose/diagnosis , Guanfacine/poisoning , Heart Failure/chemically induced , Pulmonary Edema/chemically induced , Adolescent , Drug Overdose/complications , Heart Failure/diagnosis , Humans , Male , Pulmonary Edema/diagnosis
18.
Eur J Appl Physiol ; 121(12): 3409-3419, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34480632

ABSTRACT

PURPOSE: Bolus intravenous administration of 0.9% saline has been associated with the development of pulmonary edema, and increased mortality. An animal model has previously demonstrated that rapid intravenous administration of 0.9% saline was associated with non-hydrostatic lung injury with increased lung lavage protein. We hypothesized that this non-hydrostatic effect would also occur in human volunteers. METHODS: In a randomized, cross-over study of 14 healthy male subjects, the lung lavage protein concentration and cardiorespiratory effects of an intervention with rapid intravenous administration of 30 mL/kg of 0.9% saline were compared with sham intervention. Bronchoalveolar lavage (BAL) was performed after fluid administration. Doppler echocardiography, lung ultrasound, pulmonary function tests, and blood sampling were performed before and after each intervention. RESULTS: The BAL total protein concentration was greater after 0.9% saline administration than with sham (196.1 µg/mL (SD 87.6) versus 129.8 µg/mL (SD 55.4), respectively (p = 0.020). Plasma angiopoietin-2 concentration was also increased to 2.26 ng/mL (SD 0.87) after 0.9% saline administration compared with sham 1.53 ng/mL (SD 0.69) (p < 0.001). There were small increases in stroke volume (from 58 mL (IQR 51-74) to 66 mL (IQR 58-74), p = 0.045) and Doppler echocardiography left ventricle E/e' ratio (from 5.0 (IQR 4.5-6.0) to 5.7 (IQR 5.3-6.3), p = 0.007), but no changes to right ventricular function. CONCLUSION: Rapid intravenous administration of 0.9% saline leads to interstitial permeability pulmonary edema in healthy human volunteers. Further research is now warranted to understand these effects in critically ill patients.


Subject(s)
Pulmonary Edema/chemically induced , Saline Solution/administration & dosage , Saline Solution/adverse effects , Adult , Biomarkers/blood , Bronchoalveolar Lavage , Cross-Over Studies , Echocardiography, Doppler , Healthy Volunteers , Humans , Injections, Intravenous , Male , Permeability , Prospective Studies , Respiratory Function Tests , Urinalysis
19.
J Vet Pharmacol Ther ; 44(4): 478-502, 2021 Jul.
Article in English | MEDLINE | ID: mdl-33709435

ABSTRACT

Alpha2 -adrenergic agonists have been implicated in the development of pulmonary edema (PE) and sustained hypoxemia that lead to life-threatening pulmonary distress in ruminants, especially with sensitive and compromised animals. Recently, there is limited understanding of exact mechanism underlying pulmonary alterations associated with α2 -adrenergic agonist administration. Ruminants have a rich population of pulmonary intravascular macrophages (PIMs) in the pulmonary circulation, which may be involved in the development of pulmonary alveolo-capillary barrier damage. Hence, the central thesis of this review is overviewing the literatures regarding the systemic use of α2 -adrenergic agonists in domestic ruminants, focusing on their pulmonary side effects, especially on the influence of PIMs on the lung. At this moment, further studies are needed to provide a clear emphasis and better understanding of the potential role of PIMs in the lung pathophysiology associated with α2 -adrenergic agonists. These preliminary studies would be potentially to develop future medications and intervention targets that may be helpful to alleviate or prevent the critical striking pulmonary effects, and thereby improving the safety of α2 -agonist application in ruminants.


Subject(s)
Anesthetics , Pulmonary Edema , Adrenergic alpha-2 Receptor Agonists/adverse effects , Animals , Hypoxia/chemically induced , Hypoxia/veterinary , Macrophages , Pulmonary Edema/chemically induced , Pulmonary Edema/veterinary , Ruminants
20.
J Cell Physiol ; 235(12): 9510-9523, 2020 12.
Article in English | MEDLINE | ID: mdl-32529661

ABSTRACT

Acute respiratory distress syndrome (ARDS) is a lethal clinical syndrome characterized by damage of the epithelial barriers and accumulation of pulmonary edema fluid. Protectin conjugates in tissue regeneration 1 (PCTR1), an endogenously produced lipid mediator, are believed to exert anti-inflammatory and pro-resolution effects. PCTR1 (1 µg/kg) was injected at 8 hr after lipopolysaccharide (LPS; 14 mg/kg) administration, and the rate of pulmonary fluid clearance was measured in live rats at 1 hr after PCTR1 treatment. The primary type II alveolar epithelial cells were cultured with PCTR1 (10 nmol/ml) and LPS (1 µg/ml) for 8 hr. PCTR1 effectively improved pulmonary fluid clearance and ameliorated morphological damage and reduced inflammation of lung tissue, as well as improved the survival rate in the LPS-induced acute lung injury (ALI) model. Moreover, PCTR1 markedly increased sodium channel expression as well as Na, K-ATPase expression and activity in vivo and in vitro. In addition, PCTR1i also upregulated the expression of LYVE-1 in vivo. Besides that, BOC-2, HK7, and LY294002 blocked the promoted effect of PCTR1 on pulmonary fluid clearance. Taken together, PCTR1 upregulates sodium channels' expression via activating the ALX/cAMP/P-Akt/Nedd4-2 pathway and increases Na, K-ATPase expression and activity to promote alveolar fluid clearance. Moreover, PCTR1 also promotes the expression of LYVE-1 to recover the lymphatic drainage resulting in the increase of lung interstitial fluid clearance. In summary, these results highlight a novel systematic mechanism for PCTR1 in pulmonary edema fluid clearance after ALI/ARDS, suggesting its potential role in a therapeutic approach for ALI/ARDS.


Subject(s)
Acute Lung Injury/drug therapy , CD59 Antigens/pharmacology , Epithelial Sodium Channels/genetics , Pulmonary Edema/drug therapy , Respiratory Distress Syndrome/drug therapy , Acute Lung Injury/chemically induced , Acute Lung Injury/pathology , Alveolar Epithelial Cells/drug effects , Alveolar Epithelial Cells/pathology , Animals , Anti-Inflammatory Agents/pharmacology , Body Fluids/drug effects , CD59 Antigens/chemistry , CD59 Antigens/genetics , Cyclin-Dependent Kinase Inhibitor p16 , Disease Models, Animal , Docosahexaenoic Acids/chemistry , Docosahexaenoic Acids/pharmacology , Gene Expression Regulation/drug effects , Humans , Lipopolysaccharides/toxicity , Lung/drug effects , Lung/pathology , Phosphatidylinositol 3-Kinases/genetics , Pulmonary Alveoli/drug effects , Pulmonary Alveoli/pathology , Pulmonary Edema/chemically induced , Pulmonary Edema/pathology , Rats , Respiratory Distress Syndrome/chemically induced , Respiratory Distress Syndrome/genetics , Signal Transduction/drug effects , Sodium-Potassium-Exchanging ATPase/genetics
SELECTION OF CITATIONS
SEARCH DETAIL