Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Surgeon ; 21(4): e152-e158, 2023 Aug.
Article in English | MEDLINE | ID: mdl-36192296

ABSTRACT

BACKGROUND: Inguinal hernia repair is a commonly performed surgical procedure performed in adult males. Urinary retention following surgery is a known complication likely due to the adrenergic over-stimulation of smooth muscles in the bladder neck and prostate. This effect could potentially be mitigated by the use of alpha-blocker medications. A meta-analysis of randomized control trials (RCTs) was performed to analyse the evidence behind the use of alpha-blockers in the prevention of post-operative urinary retention (POUR). METHODS: A comprehensive search of PubMed, Embase, MedLine and Scopus was undertaken adhering to PRISMA guidelines. RCTs using alpha-blockers as a single point intervention were included. Data were analysed using a random-effects model. Risk of Bias (ROB) was assessed according to Cochrane guidelines. RESULTS: Seven RCTs including 680 patients were included. The use of alpha-blockade reduced the incidence of urinary retention requiring catheterization (OR:0.23, 95% CI:0.07-0.70, p:0.009). No serious side-effects of alpha-blockers were reported. CONCLUSION: Alpha-blockers are a safe and effective intervention to reduce the incidence of urinary retention following inguinal hernia repair surgery.


Subject(s)
Hernia, Inguinal , Urinary Retention , Adult , Humans , Male , Adrenergic alpha-Antagonists/therapeutic use , Hernia, Inguinal/surgery , Hernia, Inguinal/etiology , Herniorrhaphy/adverse effects , Herniorrhaphy/methods , Incidence , Postoperative Complications/etiology , Postoperative Complications/prevention & control , Postoperative Complications/epidemiology , Urinary Retention/etiology , Urinary Retention/prevention & control , Urinary Retention/epidemiology , Randomized Controlled Trials as Topic
2.
Eur J Pharmacol ; 928: 175095, 2022 Aug 05.
Article in English | MEDLINE | ID: mdl-35728626

ABSTRACT

Snake envenomation leads to the formation of damage-associated molecular patterns (DAMPs), which are mediated by endogenous intracellular molecules. These are recognized by pattern-recognition receptors (PRRs) and can induce sterile inflammation. AIMS: In the present study, we aim at understanding the mechanisms involved in DAMPs induced sterile inflammation to unravel the novel therapeutic strategies for treating snake bites. The potential of benzodiazepinone derivatives to act against snake venom induced inflammation has been explored in the present investigation. MAIN METHODS: Three compounds VA 17, VA 43 and PA 03 were taken from our library of synthetic compounds. Oxidative stress markers such as lipid peroxidation, superoxide and nitric oxide were measured along with the analysis of DAMPs (IL6, HMGB1, vWF, S100b and HSP70). These compounds have been docked using molecular docking against the snake venom PLA2 structure (PDB code: 1OXL). KEY FINDINGS: The compounds have been found to effectively neutralize viper and cobra venoms induced lethal activity both ex vivo and in vivo. The compounds have also neutralized the viper venom induced hemorrhagic, coagulant, anticoagulant reactions as well as inflammation. The fold of protection have always been found to be higher in case of ex vivo than in in vivo. These compounds have neutralized the venom induced DAMPs as exhibited by IL6, HMGB1, vWF, S100b and HSP70. The fold of neutralization is found to be higher in VA 43. SIGNIFICANCE: The identified compounds could be used as potential candidates for developing treatment of snakebites in areas where antiserums are not yet available.


Subject(s)
HMGB1 Protein , Snake Bites , Animals , Antivenins/chemistry , Antivenins/pharmacology , Antivenins/therapeutic use , Inflammation/chemically induced , Inflammation/drug therapy , Interleukin-6 , Molecular Docking Simulation , Snake Bites/drug therapy , Viper Venoms , von Willebrand Factor
3.
PLoS One ; 16(12): e0260835, 2021.
Article in English | MEDLINE | ID: mdl-34882718

ABSTRACT

Myocardial infarction (MI), atherosclerosis and other inflammatory and ischemic cardiovascular diseases (CVDs) have a very high mortality rate and limited therapeutic options. Although the diagnosis is based on markers such as cardiac Troponin-T (cTrop-T), the mechanism of cTrop-T upregulation and release is relatively obscure. In the present study, we have investigated the mechanism of cTrop-T release during acute hypoxia (AH) in a mice model by ELISA & immunohistochemistry. Our study showed that AH exposure significantly induces the expression and release of sterile inflammatory as well as MI markers in a time-dependent manner. We further demonstrated that activation of TLR3 (mediated by eRNA) by AH exposure in mice induced cTrop-T release and Poly I:C (TLR3 agonist) also induced cTrop-T release, but the pre-treatment of TLR3 immuno-neutralizing antibody or silencing of Tlr3 gene or RNaseA treatment two hrs before AH exposure, significantly abrogated AH-induced Caspase 3 activity as well as cTrop-T release. Our immunohistochemistry and Masson Trichrome (MT) staining studies further established the progression of myocardial injury by collagen accumulation, endothelial cell and leukocyte activation and adhesion in myocardial tissue which was abrogated significantly by pre-treatment of RNaseA 2 hrs before AH exposure. These data indicate that AH induced cTrop-T release is mediated via the eRNA-TLR3-Caspase 3 pathway.


Subject(s)
Apoptosis , Extracellular Vesicles/genetics , Hypoxia/physiopathology , Myocardial Infarction/pathology , RNA/genetics , Toll-Like Receptor 3/metabolism , Troponin T/metabolism , Animals , Caspase 3/genetics , Caspase 3/metabolism , Disease Models, Animal , Mice , Myocardial Infarction/genetics , Myocardial Infarction/metabolism , RNA/metabolism , Signal Transduction , Toll-Like Receptor 3/genetics
4.
Blood Cells Mol Dis ; 84: 102459, 2020 09.
Article in English | MEDLINE | ID: mdl-32559654

ABSTRACT

Sterile Inflammation (SI), a condition where damage associated molecular patterns (DAMPs) released from dying cells, leads to TLR (Toll-like receptor) activation and triggers hypoxemia in circulation leading to venous thrombosis (VT) through tissue factor (TF) activation, but its importance under acute hypoxia (AH) remains unexplored. Thus, we hypothesized that eRNA released from dying cells under AH activates TF via the TLR3-ERK1/2-AP1 pathway, leading to VT. Animals were exposed to stimulate hypoxia for 0-24 h at standard temperature and humidity. RNaseA and DNase1 were injected immediately before exposure. TLR3 gene silencing was performed through in vivo injection of TLR3 siRNA. 80 µg/kg BW of isolated eRNA and eDNA were injected 6 h prior to sacrifice. Antigens of TF pathway were determined by ELISA and TF activity by a chromogenic assay. AH exposure significantly induced release of SI markers i.e. eRNA, eDNA, HMGB1 and upregulated TLR3, ERK1/2 (Extracellular signal-regulated kinases), AP1 (Activator Protein-1) and TF, whereas RNaseA pre-treatment diminished the effect of AH, thus inhibiting TF expression as well as activity during AH. Hence, we propose a possible mechanism of AH-induced TF activation and thrombosis where RNaseA can become the novel focal point in ameliorating therapy for AH induced thrombosis.


Subject(s)
Hypoxia/metabolism , RNA/metabolism , Replication Protein C/metabolism , Signal Transduction , Thromboplastin/metabolism , Toll-Like Receptor 3/metabolism , Animals , Cells, Cultured , Hypoxia/complications , Hypoxia/genetics , MAP Kinase Signaling System , Mice , Thromboplastin/genetics , Thrombosis/etiology , Thrombosis/genetics , Thrombosis/metabolism , Up-Regulation
5.
Sci Rep ; 9(1): 11175, 2019 08 01.
Article in English | MEDLINE | ID: mdl-31371738

ABSTRACT

Sterile inflammation (SI) is an essential process in response to snakebite and injury. The venom induced pathophysiological response to sterile inflammation results into many harmful and deleterious effects that ultimately leads to death. The available treatment for snakebite is antiserum which does not provide enough protection against venom-induced pathophysiological changes like haemorrhage, necrosis, nephrotoxicity and often develop hypersensitive reactions. In order to overcome these hindrances, scientists around the globe are searching for an alternative therapy to provide better treatment to the snake envenomation patients. In the present study TiO2 (Titanium dioxide)-NPs (Nanoparticles) has been assessed for antisnake venom activity and its potential to be used as an antidote. In this study, the synthesis of TiO2-NPs arrays has been demonstrated on p-type Silicon Si < 100 > substrate (∼30 ohm-cm) and the surface topography has been detected by Field-emission scanning electron microscopy (FESEM). The TiO2-NPs successfully neutralized the Daboia russelii venom (DRV) and Naja kaouthia venom (NKV)-induced lethal activity. Viper venom induced haemorrhagic, coagulant and anticoagulant activities were effectively neutralized both in in-vitro and in vivo studies. The cobra and viper venoms-induced sterile inflammatory molecules (IL-6, HMGB1, HSP70, HSP90, S100B and vWF) were effectively neutralised by the TiO2-NPs in experimental animals.


Subject(s)
Inflammation/prevention & control , Nanoparticles/therapeutic use , Phospholipases A2/metabolism , Snake Venoms/antagonists & inhibitors , Titanium/therapeutic use , Animals , Animals, Laboratory , Elapid Venoms , Hemorrhage/prevention & control , Mice , Nanoparticles/chemistry , Necrosis/prevention & control , Snake Bites/pathology , Snake Bites/physiopathology , Snake Bites/therapy , Snake Venoms/toxicity , Viper Venoms
6.
J Leukoc Biol ; 105(4): 705-717, 2019 04.
Article in English | MEDLINE | ID: mdl-30835912

ABSTRACT

PGE2 plays a critical role in angiogenesis, ischemic, and neuro-inflammatory disorders of the brain, which breakdown the blood-brain barrier (BBB). However, the effects of PGE2 on human brain endothelial cell (HBECs) migration, a key process in the angiogenic response and BBB stability, are not well defined. In this study, we investigated the mechanism of PGE2 in HBECs migration in vitro. Here we showed that PGE2 stimulated migration of HBECs in a dose-time and matrix-dependent manner, evaluated by the Boyden chamber assay, but other prostanoids failed to do so. PGE2 receptor (EP2; butaprost), EP3 (sulprostone), and EP4 (PGE1 -OH) receptor agonists stimulated HBECs migration, but the silencing of EP significantly attenuated this effect. EP1 agonist (11-trinor PGE1 ) had no effect on HBECs migration on silencing of the EP1 receptor. We further showed that PGE2 stimulated cAMP production and activated protein kinase A (PKA), whereas pretreatment with the adenyl cyclase inhibitor (dideoxyadenosine; 1 µM) or PKA inhibitors, H89 (0.5 µM)/PKAI (1 µM), completely abrogated PGE2 -induced migration. Furthermore, silencing of the EP2/EP4 receptors significantly inhibited PGE2 -induced cAMP and PKA activation, whereas EP3 receptor silencing failed to do so. These results suggest that PGE2 regulates HBEC migration via cooperation of EP2, EP3, and EP4 receptors. Coupling of PGE2 to these receptors resulted in increased production of cAMP, which regulates HBEC migration via PKA pathway. The elucidation of molecular events involved is critical for the development of targeted strategies to treat cerebrovascular diseases associated with dysregulated angiogenesis.


Subject(s)
Brain/cytology , Cell Movement , Cyclic AMP-Dependent Protein Kinases/metabolism , Dinoprostone/pharmacology , Endothelial Cells/cytology , Endothelial Cells/metabolism , Receptors, Prostaglandin E/metabolism , Cell Movement/drug effects , Chemotaxis/drug effects , Cyclic AMP/biosynthesis , Endothelial Cells/drug effects , Extracellular Matrix/drug effects , Extracellular Matrix/metabolism , Humans
7.
J Vasc Res ; 55(4): 244-254, 2018.
Article in English | MEDLINE | ID: mdl-30223272

ABSTRACT

High mobility group box 1 protein (HMGB1), a sterile inflammatory molecule and damage-associated molecular pattern (DAMP) released from various cells during stress has been implicated in inflammation. Several reports show that there is a direct relationship between inflammation and cardiovascular diseases (CVDs) such as thrombosis, hypertension, insulin resistance, preeclampsia, etc. Here, we intend to summarize the concept of the emerging link between HMGB1 and CVDs. Furthermore, we will discuss the possible therapeutic strategies that target HMGB1 for the treatment of different CVDs.


Subject(s)
Cardiovascular Diseases/physiopathology , HMGB1 Protein/physiology , Inflammation/physiopathology , Animals , Cardiovascular Diseases/drug therapy , Coronary Disease/physiopathology , Disseminated Intravascular Coagulation/physiopathology , Female , HMGB1 Protein/antagonists & inhibitors , Humans , Oxidation-Reduction , Pre-Eclampsia/physiopathology , Pregnancy , Signal Transduction , Stroke/physiopathology , Venous Thrombosis/physiopathology
8.
Eur J Immunol ; 46(10): 2388-2400, 2016 10.
Article in English | MEDLINE | ID: mdl-27480067

ABSTRACT

Increased plasma level of von Willebrand Factor (vWF) is associated with major cardiovascular diseases. We previously reported that multimeric vWF binds to NO synthase and inhibits insulin-induced production of NO, thus promoting insulin resistance during acute hypoxia (AH). However, the transcriptional regulation of vWF during AH is not clearly understood. Here, we investigated the mechanisms underlying the upregulation of vwf in mice. AH significantly upregulates the tlr2, tlr3, myd88, and vwf expression and phosphorylation of specificity protein 1 (SP1). Furthermore, AH significantly upregulates high mobility group box-1 (HMGB1) in a time-dependent manner. Moreover, a TLR2 agonist upregulates vWF but a TLR3 agonist does not. Pretreatment with an HMGB1 inhibitor, TLR2-immunoneutralizing antibody, or SP1 inhibitor significantly inhibits vWF expression. Furthermore, Tlr2 silencing completely inhibited MYD88, vWF expression, and SP1 phosphorylation. However, pretreatment with glycyrrhizic acid or silencing of Tlr2 completely blocks binding of Sp1 to the Vwf promoter, thus inhibiting its expression, and enhances insulin resistance during AH. Patients with type 2 diabetes mellitus also showed significantly elevated levels of HMGB1, TLR2, SP1, and vWF, thereby supporting the results of the murine model of AH. Taken together, HMGB1 upregulates vWF in vivo through the TLR2-MYD88-SP1 pathway in mice.


Subject(s)
Diabetes Mellitus, Type 2/immunology , HMGB1 Protein/metabolism , Hypoxia/immunology , von Willebrand Factor/metabolism , Animals , Antibodies, Blocking/administration & dosage , Cells, Cultured , Diabetes Mellitus, Type 2/therapy , Glycyrrhizic Acid/pharmacology , HMGB1 Protein/genetics , Humans , Hypoxia/therapy , Immunoglobulins/metabolism , Insulin Resistance , Mice , Molecular Targeted Therapy , Myeloid Differentiation Factor 88/metabolism , RNA, Small Interfering/genetics , Signal Transduction , Toll-Like Receptor 2/genetics , Toll-Like Receptor 2/metabolism , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL
...