Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Eur J Pharmacol ; 974: 176633, 2024 Jul 05.
Article in English | MEDLINE | ID: mdl-38703975

ABSTRACT

Cardiac arrest (CA) remains a leading cause of death, with suboptimal survival rates despite efforts involving cardiopulmonary resuscitation and advanced life-support technology. Post-resuscitation myocardial dysfunction (PRMD) is an important determinant of patient outcomes. Myocardial ischemia/reperfusion injury underlies this dysfunction. Previous reports have shown that ruthenium red (RR) has a protective effect against cardiac ischemia-reperfusion injury; however, its precise mechanism of action in PRMD remains unclear. This study investigated the effects of RR on PRMD and analyzed its underlying mechanisms. Ventricular fibrillation was induced in rats, which were then subjected to cardiopulmonary resuscitation to establish an experimental CA model. At the onset of return of spontaneous circulation, RR (2.5 mg/kg) was administered intraperitoneally. Our study showed that RR improved myocardial function and reduced the production of oxidative stress markers such as malondialdehyde (MDA), glutathione peroxidase (GSSG), and reactive oxygen species (ROS) production. RR also helped maintain mitochondrial structure and increased ATP and GTP levels. Additionally, RR effectively attenuated myocardial apoptosis. Furthermore, we observed downregulation of proteins closely related to mitophagy, including ubiquitin-specific protease 33 (USP33) and P62, whereas LC3B (microtubule-associated protein light chain 3B) was upregulated. The upregulation of mitophagy may play a critical role in reducing myocardial injury. These results demonstrate that RR may attenuate PRMD by promoting mitophagy through the inhibition of USP33. These effects are likely mediated through diverse mechanisms, including antioxidant activity, apoptosis suppression, and preservation of mitochondrial integrity and energy metabolism. Consequently, RR has emerged as a promising therapeutic approach for addressing post-resuscitation myocardial dysfunction.


Subject(s)
Disease Models, Animal , Heart Arrest , Mitophagy , Rats, Sprague-Dawley , Ruthenium Red , Animals , Mitophagy/drug effects , Heart Arrest/complications , Heart Arrest/drug therapy , Heart Arrest/metabolism , Heart Arrest/physiopathology , Rats , Male , Ruthenium Red/pharmacology , Ruthenium Red/therapeutic use , Oxidative Stress/drug effects , Ubiquitin Thiolesterase/metabolism , Cardiopulmonary Resuscitation , Up-Regulation/drug effects , Myocardium/pathology , Myocardium/metabolism , Apoptosis/drug effects , Reactive Oxygen Species/metabolism , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/drug therapy , Myocardial Reperfusion Injury/physiopathology
2.
Biochem Biophys Res Commun ; 635: 236-243, 2022 12 20.
Article in English | MEDLINE | ID: mdl-36283336

ABSTRACT

Acute pancreatitis (AP) is a common inflammatory disease of the digestive system. Mitochondrial calcium uniporter (MCU) mediates mitochondrial uptake of Ca2+ and plays an important role in calcium homeostasis. However, it is undefined whether AP can be relieved by inhibiting MCU. This study aimed to study the therapeutic potential of Ruthenium red (RuR), a MCU inhibitor, in AP mice model and primary acinar cells. Cell injury and AP mice model was induced by caerulein. RuR alleviated CER-AP evidenced by reduced serum lipase, TNF-α, and pancreatic MPO levels, less severe pancreatic pathology damage, and decreased inflammatory cell infiltration. In freshly isolated pancreatic acinar cells, RuR diminished cell necrosis with effect on suppressing the expression of MCU. RuR also decreased levels of cytosolic calcium and ROS, preventing mitochondrial membrane potential loss, ATP depletion and MPTP opening. The present findings indicate that inhibit MCU by RuR has a beneficial effect in AP by preventing calcium overload, mitochondrial dysfunction, and cell necrosis.


Subject(s)
Calcium Channel Blockers , Calcium , Pancreatitis , Ruthenium Red , Animals , Mice , Acute Disease , Calcium/metabolism , Mitochondria/metabolism , Necrosis/pathology , Pancreatitis/drug therapy , Pancreatitis/pathology , Ruthenium Red/therapeutic use , Calcium Channel Blockers/therapeutic use
3.
Oxid Med Cell Longev ; 2022: 7727006, 2022.
Article in English | MEDLINE | ID: mdl-36148414

ABSTRACT

Osteoporosis is a disorder of bone metabolism that is extremely common in elderly patients as well as in postmenopausal women. The main manifestation is that the bone resorption capacity is greater than the bone formation capacity, which eventually leads to a decrease in bone mass, increasing the risk of fracture. There is growing evidence that inhibiting osteoclast formation and resorption ability can be effective in treating and preventing the occurrence of osteoporosis. Our study is the first time to explore the role of the mitochondrial calcium uniporter (MCU) and its inhibitor ruthenium red (RR) in bone metabolism, clarifying the specific mechanism by which it inhibits osteoclast formation in vitro and plays a therapeutic role in osteoporosis in vivo. We verified the suppressive effects of RR on the receptor activator of nuclear factor-κB ligand (RANKL-)-induced differentiation and bone resorption function of osteoclasts in vitro. The reactive oxygen species (ROS) production stimulated by RANKL and the expression level of P38 MAPK/NFATc1 were also found to be inhibited by RR. Moreover, the promotion of RR on osteogenesis differentiation was investigated by alkaline phosphatase (ALP) and alizarin red S (ARS) staining and the detection of osteogenesis-specific gene expression levels by quantitative polymerase chain reaction (qPCR) and western blotting. Moreover, in ovariectomy (OVX-)-induced osteoporosis models, RR can downregulate the expression and function of the MCU, relieving bone loss and promoting osteogenesis to present a therapeutic effect on osteoporosis. This new finding will provide an important direction for the study of RR and MCU in the study of bone metabolism therapy targets.


Subject(s)
Bone Resorption , Osteoporosis , Aged , Alkaline Phosphatase/genetics , Bone Resorption/drug therapy , Bone Resorption/metabolism , Calcium Channels , Cell Differentiation , Female , Gene Expression , Humans , NFATC Transcription Factors , Osteoclasts/metabolism , Osteogenesis , Osteoporosis/drug therapy , Osteoporosis/metabolism , Ovariectomy , RANK Ligand/metabolism , Reactive Oxygen Species/metabolism , Ruthenium Red/metabolism , Ruthenium Red/pharmacology , Ruthenium Red/therapeutic use , Signal Transduction , p38 Mitogen-Activated Protein Kinases/metabolism
4.
Eur J Pharmacol ; 902: 174113, 2021 Jul 05.
Article in English | MEDLINE | ID: mdl-33901460

ABSTRACT

The transient receptor potential vanilloid channel 4 (TRPV4) is associated with the development of several pathologies, particularly gastric disorders. However, there are no studies associating this receptor with the pathophysiology of gastric erosions. The aim of this study was to investigate the role of TRPV4 in the development of ethanol-induced gastric damage in vivo. Gastric lesions were induced by ethanol in Swiss mice pretreated with TRPV4 antagonists, GSK2193874 (0.1; 0.3 and 0.9 mg/kg) or Ruthenium red (0.03; 0.1 or 0.3 mg/kg) or its agonist, GSK1016790A (0.9 mg/kg). Gastric mucosal samples were taken for histopathology, immunohistochemistry, atomic force microscopy and evaluation of antioxidant parameters. The gastric mucus content and TRPV4 mRNA expression were analyzed. Ethanol exposure induced upregulation of gastric mRNA and protein expression of TRPV4. TRPV4 blockade promoted gastroprotection against ethanol-induced injury on macro- and microscopic levels, leading to reduced hemorrhage, cell loss and edema and enhanced gastric mucosal integrity. Moreover, an increase in superoxide dismutase (SOD) and glutathione (GSH) activity was observed, followed by a decrease in malondialdehyde (MDA) levels. TRPV4 blockade during alcohol challenge reestablished gastric mucus content. The combination of TRPV4 agonist and ethanol revealed macroscopic exacerbation of gastric damage area. Our results confirmed the association of TRPV4 with the development of gastric injury, showing the importance of this receptor for further investigations in the field of gastrointestinal pathophysiology and pharmacology.


Subject(s)
Stomach Ulcer/metabolism , Stomach Ulcer/physiopathology , TRPV Cation Channels/agonists , TRPV Cation Channels/metabolism , Animals , Edema/chemically induced , Edema/metabolism , Ethanol/toxicity , Gastric Mucosa/drug effects , Gastric Mucosa/injuries , Gastric Mucosa/metabolism , Glutathione/metabolism , Leucine/analogs & derivatives , Leucine/pharmacology , Leucine/therapeutic use , Male , Malondialdehyde/metabolism , Mice , Oxidative Stress/drug effects , Piperidines/pharmacology , Piperidines/therapeutic use , Quinolines/pharmacology , Quinolines/therapeutic use , Ruthenium Red/pharmacology , Ruthenium Red/therapeutic use , Stomach Ulcer/chemically induced , Stomach Ulcer/pathology , Sulfonamides/pharmacology , Sulfonamides/therapeutic use , Superoxide Dismutase/metabolism , TRPV Cation Channels/antagonists & inhibitors , TRPV Cation Channels/genetics , Up-Regulation/drug effects
5.
Brain Res Bull ; 164: 121-135, 2020 11.
Article in English | MEDLINE | ID: mdl-32858127

ABSTRACT

Alzheimer's disease (AD) is a progressive neurodegenerative disorder with cardinal features of cognitive dysfunction in an individual. Recently, the blockade of mitochondrial calcium uniporter (MCU) exhibits neuroprotective activity in experimental animals. However, the therapeutic potential of MCU has not yet been established in the management of AD. Therefore, the present study explored the therapeutic potential of either Ruthenium red (RR), a MCU blocker, or Spermine, a MCU opener, on the extent of mitochondrial calcium accumulation, function, integrity and bioenergetics in hippocampus, pre-frontal cortex and amygdale of ICV-STZ challenged rats. Experimental AD was induced in male rats by intracerebroventricular injection of streptozotocin (ICV-STZ) on day-1 (D-1) of the experimental protocol at a sub-diabetogenic dose (3 mg/kg) twice at an interval of 48 h into both rat lateral ventricles. RR attenuated ICV-STZ-induced memory-related behavioral abnormalities in Morris water maze and Y-maze tests. RR also attenuated ICV-STZ-induced decrease in the level of acetylcholine and activity of choline acetyltransferase and, increase in the activity of acetylcholinestarase in memory-sensitive rat brain regions. Further, RR attenuated mitochondrial toxicity in terms of reducing mitochondrial calcium accumulation and improving the mitochondrial function, integrity and bioenergetics in memory-sensitive brain regions of ICV-STZ challenged rats. Furthermore, RR attenuated the percentage of apoptotic cells in ICV-STZ challenged rat brain regions. However, Spermine did not alter ICV-STZ-induced behavioral, biochemical and molecular observations in any of the brain regions. These observations indicate the fact that the MCU blockage could be a potential therapeutic option in the management of sporadic type of AD.


Subject(s)
Calcium Channels/pharmacology , Cognition Disorders/drug therapy , Cognition/drug effects , Maze Learning/drug effects , Neuroprotective Agents/pharmacology , Ruthenium Red/pharmacology , Acetylcholine/metabolism , Acetylcholinesterase/metabolism , Animals , Calcium Channels/therapeutic use , Cognition Disorders/chemically induced , Cognition Disorders/metabolism , Male , Mitochondria/drug effects , Mitochondria/metabolism , Neuroprotective Agents/therapeutic use , Rats , Rats, Sprague-Dawley , Ruthenium Red/therapeutic use , Streptozocin
6.
Int J Cardiol ; 271: 161-168, 2018 Nov 15.
Article in English | MEDLINE | ID: mdl-29803339

ABSTRACT

BACKGROUND: HF incurs high disease burden, and the effectiveness of known HF treatments is unsatisfactory. Therefore, seeking novel therapeutic target of HF is important. The present study aimed to investigate the role of the mitochondrial calcium uniporter (MCU) and its relationship with autophagy in overload-induced heart failure (HF). METHODS AND RESULTS: In both early-stage and end-stage of pressure overload-induced HF, MCU appeared up-regulated along with heart enlargement, increased microtubule-associated proteins 1A/1B light chain 3B (LC3B) II/I ratio and autophagosome content, damaged cardiac function, and ventricular asynchrony. However, sequestosome-1 (SQSTM1/p62) level decreased indicating blockaded autophagic flux. Seven-week administration of MCU inhibitor ruthenium red improved cardiac function and mitigated its pathological change. MCU inhibition maintained mitochondrial integrity, increased LC3B II/I ratio, up-regulated Parkin and Pink1, and down-regulated SQSTM1/p62. MCU inhibition also alleviated ventricular asynchrony of HF, and this might be related to connexin-43 up-regulation. In vitro study validated intervention on MCU leading to elevation of autophagy and mitophagy. MCU inhibition could partly prevent from excessive cellular enlargement induced by isoprenaline. CONCLUSIONS: In summary, MCU inhibition played an important role in pressure overload-induced heart failure through autophagy and mitophagy enhancement, and intervention on MCU offered cardioprotective effects. To our knowledge, the role of MCU in HF and its relationship with autophagy and mitophagy are firstly disclosed. Moreover, our study suggests that MCU inhibition could be explored as a novel therapeutic concept in HF.


Subject(s)
Autophagy/physiology , Calcium Channels/biosynthesis , Heart Failure/metabolism , Heart Failure/prevention & control , Ruthenium Red/pharmacology , Animals , Autophagy/drug effects , Cells, Cultured , Heart Failure/diagnostic imaging , Male , Mice , Mice, Inbred C57BL , Mitochondria, Heart/drug effects , Mitochondria, Heart/metabolism , Ruthenium Red/therapeutic use , Up-Regulation/drug effects , Up-Regulation/physiology
7.
Cell Physiol Biochem ; 46(2): 687-698, 2018.
Article in English | MEDLINE | ID: mdl-29621761

ABSTRACT

BACKGROUND/AIMS: Chondrocyte apoptosis is the most common pathological feature in cartilage in osteoarthritis (OA). Transient receptor potential channel vanilloid 5 (TRPV5) is important in regulating calcium ion (Ca2+) influx. Accumulating evidences suggest that Ca2+ is a major intracellular second messenger that can trigger cell apoptosis. Therefore, we investigate the potential role of TRPV5 in mediating Ca2+ influx to promote chondrocyte apoptosis in OA. METHODS: The monoiodoacetic acid (MIA)-induced rat OA model was assessed by macroscopic and radiographic analyses. Calmodulin protein immunolocalization was detected by immunohistochemistry. The mRNA and protein level of TRPV5, calmodulin and cleaved caspase-8 in articular cartilage were assessed by real time polymerase chain reaction and western blotting. Primary chondrocytes were isolated and cultured in vitro. TRPV5 small interfering RNA was used to silence TRPV5 in chondrocytes. Then, calmodulin and cleaved caspase-8 were immunolocalized by immunofluorescence in chondrocyte. Fluo-4AM staining was used to assess intracellular Ca2+ to reflect TRPV5 function of mediation Ca2+ influx. Annexin V-fluorescein isothiocyanatepropidium iodide flow cytometric analysis was performed to determine chondrocytes apoptosis. Western blotting techniques were used to measure the apoptosis-related proteins in chondrocyte level. RESULTS: Here, we reported TRPV5 was up-regulated in MIA-induced OA articular cartilage. Ruthenium red (a TRPV5 inhibitor) can relieve progression of joint destruction in vivo which promoted us to demonstrate the effect of TRPV5 in OA. We found that TRPV5 had a specific role in mediating extracellular Ca2+ influx leading to chondrocytes apoptosis in vitro. The apoptotic effect was inhibited even reversed by silencing TRPV5. Furthermore, we found that the increase Ca2+ influx triggered apoptosis by up-regulating the protein of death-associated protein, FAS-associated death domain, cleaved caspase-8, cleaved caspase-3, cleaved caspase-6, and cleaved caspase-7, and the up-regulated proteins were abolished by silencing TRPV5 or 1, 2-bis-(o-Aminophenoxy)-ethane-N,N,N',N'-tetraacetic acid, tetraacetoxymethyl ester (a Ca2+ chelating agent). CONCLUSION: The up-regulated TRPV5 could used be as an initiating factor that induces extrinsic chondrocyte apoptosis via the mediation of Ca2+ influx. These findings suggested TRPV5 could be an intriguing mediator for drug target in OA.


Subject(s)
Apoptosis , Calcium Channels/metabolism , Calcium/metabolism , Osteoarthritis/pathology , TRPV Cation Channels/metabolism , Animals , Apoptosis/drug effects , Calcium Channels/genetics , Calcium Chelating Agents/pharmacology , Calmodulin/genetics , Calmodulin/metabolism , Cartilage, Articular/metabolism , Cartilage, Articular/pathology , Caspase 3/metabolism , Caspase 7/metabolism , Caspase 8/genetics , Caspase 8/metabolism , Cells, Cultured , Chondrocytes/cytology , Chondrocytes/metabolism , Disease Models, Animal , Immunohistochemistry , Iodoacetic Acid/toxicity , Male , Osteoarthritis/chemically induced , Osteoarthritis/drug therapy , RNA Interference , RNA, Small Interfering/metabolism , Rats , Rats, Sprague-Dawley , Ruthenium Red/pharmacology , Ruthenium Red/therapeutic use , TRPV Cation Channels/antagonists & inhibitors , TRPV Cation Channels/genetics , Up-Regulation/drug effects
8.
J Pharmacol Sci ; 133(4): 254-260, 2017 Apr.
Article in English | MEDLINE | ID: mdl-28410966

ABSTRACT

The clinical anti-cancer efficacy of vincristine is limited by the development of dose-dependent peripheral neuropathy. Up-regulation of transient receptor potential vanilloid 1 (TRPV1) is correlated with peripheral neuropathy following anti-cancer drug treatment. To analyze the contribution of TRPV1 to the development of vincristine-induced mechanical allodynia/hyperalgesia, TRPV1 expression in the rat dorsal root ganglion (DRG) was analyzed after vincristine treatment. Mechanical allodynia/hyperalgesia was tested with von Frey filaments 14 days after intraperitoneal administration of 0.1 mg/kg vincristine in rats. TRPV1 expression in DRGs following vincristine treatment was assessed with western blot analysis and in situ hybridization histochemistry. Vincristine-induced mechanical allodynia/hyperalgesia after day 14 was significantly inhibited by the TRP antagonist ruthenium red (3 mg/kg, s.c.) and the TRPV1 antagonist capsazepine (30 mg/kg, s.c.). Vincristine treatment increased the expression of TRPV1 protein in DRG neurons. In situ hybridization histochemistry revealed that most of the TRPV1 mRNA-labeled neurons in the DRG were small in size. Immunohistochemistry showed that isolectin B4-positive small DRG neurons co-expressed TRPV1 protein 14 days after treatment. These results suggest that vincristine treatment increases TRPV1 expression in small DRG neurons. TRPV1 expression may contribute to the development of vincristine-induced painful peripheral neuropathy.


Subject(s)
Antineoplastic Agents, Phytogenic/toxicity , Gene Expression/drug effects , Neuralgia/chemically induced , Neuralgia/genetics , TRPV Cation Channels/genetics , TRPV Cation Channels/metabolism , Vincristine/toxicity , Animals , Capsaicin/analogs & derivatives , Capsaicin/pharmacology , Capsaicin/therapeutic use , Disease Models, Animal , Dose-Response Relationship, Drug , Ganglia, Spinal/cytology , Ganglia, Spinal/metabolism , Ganglia, Spinal/pathology , Humans , Male , Neuralgia/drug therapy , Neurons/metabolism , Neurons/pathology , Rats, Wistar , Ruthenium Red/pharmacology , Ruthenium Red/therapeutic use , TRPV Cation Channels/antagonists & inhibitors , Up-Regulation/drug effects
9.
Physiol Behav ; 164(Pt A): 140-50, 2016 10 01.
Article in English | MEDLINE | ID: mdl-27262216

ABSTRACT

Diabetes mellitus is considered as a main risk factor for vascular dementia. In the past, we have reported the induction of vascular dementia by experimental diabetes. This study investigates the efficacy of a ruthenium red, a ryanodine receptor antagonist and pioglitazone in the pharmacological interdiction of pancreatectomy diabetes (PaD) induced vascular endothelial dysfunction and subsequent vascular dementia in rats. Attentional set shifting and Morris water-maze test were used for assessment of learning and memory. Vascular endothelial function, blood brain barrier permeability, serum glucose, serum nitrite/nitrate, oxidative stress (viz. aortic superoxide anion, brain thiobarbituric acid reactive species and brain glutathione), brain calcium and inflammation (myeloperoxidase) were also estimated. PaD rats have shown impairment of endothelial function, blood brain barrier permeability, learning and memory along with an increase in brain inflammation, oxidative stress and calcium. Administration of ruthenium red and pioglitazone has significantly attenuated PaD induced impairment of learning, memory, blood brain barrier permeability, endothelial function and biochemical parameters. It may be concluded that ruthenium red, a ryanodine receptor antagonist and pioglitazone, a PPAR-γ agonist may be considered as potent pharmacological agent for the management of PaD induced endothelial dysfunction and subsequent vascular dementia. Ryanodine receptor may be explored further for their possible benefits in vascular dementia.


Subject(s)
Calcium Channel Blockers/therapeutic use , Dementia/etiology , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Experimental/drug therapy , Endothelial Cells/pathology , Ruthenium Red/therapeutic use , Animals , Aorta/metabolism , Aorta/pathology , Blood Glucose/metabolism , Body Weight/drug effects , Body Weight/physiology , Brain/metabolism , Diabetes Mellitus, Experimental/etiology , Disease Models, Animal , Male , Maze Learning/drug effects , Nitrates/blood , Nitrites/blood , Pancreatectomy/adverse effects , Pioglitazone , Rats , Rats, Wistar , Reaction Time/drug effects , Ryanodine Receptor Calcium Release Channel/metabolism , Thiazolidinediones/therapeutic use
10.
Curr Neurovasc Res ; 13(1): 10-21, 2016.
Article in English | MEDLINE | ID: mdl-26500103

ABSTRACT

Chronic cerebral hypoperfusion (CCH) is a general pathophysiological condition occurring in vascular dementia (VaD) associated with negative impact on cognitive functions. Ryanodine as well as cysteinyl leukotriene-1 receptors (RyRs and CysLT1Rs) are extensively present in the central nervous system, where they participate in regulation of cognition, motivation, inflammation and neurodegeneration. The purpose of this study is to examine the role of ruthenium red; a selective RyR blocker as well as montelukast; a specific CysLT1 antagonist in CCH induced VaD in mice. Two vessel occlusion (2VO) or permanent ligation of bilateral common carotid arteries technique was used to induce CCH in mice. Animals with bilateral carotid arteries occlusion have revealed impaired learning and memory (Morris water maze), cholinergic dysfunction (increased acetylcholinesterase activity) as well as increased brain oxidative stress (reduction in brain superoxide dismutase, glutathione and catalase with an increase in thiobarbituric acid reactive substance level), with increased brain infarct size (2,3,5-triphenylterazolium chloride staining). While, administration of ruthenium red and montelukast considerably attenuated CCH induced cognitive impairments, cholinergic dysfunction, brain oxidative stress as well as brain damage. The results suggest that bilateral carotid arteries occlusion induced CCH has brought out VaD, which was attenuated by treatment with ruthenium red and montelukast. Therefore, modulation of RyRs as well as CysLT1 receptors may provide help in conditions involving CCH such as cognitive impairment and VaD.


Subject(s)
Carotid Artery Diseases , Cerebral Cortex/physiopathology , Cholinergic Agents/metabolism , Cognition Disorders/etiology , Oxidative Stress/physiology , Receptors, Leukotriene/metabolism , Ryanodine Receptor Calcium Release Channel/metabolism , Acetates/pharmacology , Acetates/therapeutic use , Animals , Carotid Artery Diseases/complications , Carotid Artery Diseases/metabolism , Carotid Artery Diseases/pathology , Cerebral Cortex/drug effects , Cognition Disorders/drug therapy , Cyclopropanes , Disease Models, Animal , Glutathione/metabolism , Leukotriene Antagonists/pharmacology , Leukotriene Antagonists/therapeutic use , Lipid Peroxidation/drug effects , Male , Mice , Oxidative Stress/drug effects , Oxidoreductases/metabolism , Quinolines/pharmacology , Quinolines/therapeutic use , Ruthenium Red/pharmacology , Ruthenium Red/therapeutic use , Sulfides , Time Factors
11.
Neurosci Lett ; 495(3): 173-7, 2011 May 20.
Article in English | MEDLINE | ID: mdl-21315136

ABSTRACT

The present work explored the antinociceptive effects of the flavonoid myricitrin in models of overt nociception triggered by intraplantar injection of chemical algogens into the hind paw of mice. The nociception induced by bradykinin (3 nmol/paw i.pl.) was abolished by prior treatment with myricitrin (10-100mg/kg, i.p.) with ID(50) of 12.4 (8.5-18.1)mg/kg. In sharp contrast, myricitrin failed to affect the nociception elicited by prostaglandin E(2) (3 nmol/paw i.pl.). Cinnamaldehyde (10 nmol/paw i.pl.)-induced nociception was reduced by myricitrin (100mg/kg, i.p.) and camphor (7.6 mg/kg,s.c.) in 43±10% and 57±8%, respectively. Myricitrin (30-100mg/kg, i.p.) and amiloride (100mg/kg, i.p.) inhibited nociceptive responses induced by acidified saline (pH 5/paw i.pl.), with ID(50) of 22.0 (16.1-30.0)mg/kg and inhibition of 71±6% and 64±5%, respectively. Moreover, myricitrin (10-30 mg/kg, i.p.) and ruthenium red (3mg/kg, i.p.) significantly reduced the nociception induced by menthol (1.2 µmol/paw i.pl.) with the mean ID(50) of 2.4 (1.5-3.7)mg/kg and inhibition of 95±3% and 51±7%, respectively. In addition, myricitrin administration (30 and 100mg/kg, i.p.) markedly reduced menthol-induced mechanical allodynia. However, myricitrin (100mg/kg, i.p.) prevented (only in time of 60 min) cold allodynia induced by menthol. Collectively, the present results extend prior data and show that myricitrin promotes potent antinociception, an action that is likely mediated by an inhibition of the activation of nociceptors by bradykinin and TRPs agonist (i.e. cinnamaldehyde, acidified saline and menthol), probably via inhibition of PKC pathways. Thus, myricitrin could constitute an attractive molecule of interest for the development of new analgesic drugs.


Subject(s)
Analgesics, Non-Narcotic/therapeutic use , Flavonoids/therapeutic use , Models, Chemical , Pain/drug therapy , Acrolein/analogs & derivatives , Acrolein/toxicity , Amiloride/therapeutic use , Animals , Bradykinin/toxicity , Camphor/therapeutic use , Dinoprostone/toxicity , Disease Models, Animal , Dose-Response Relationship, Drug , Functional Laterality/drug effects , Hyperalgesia/drug therapy , Male , Mice , Pain/chemically induced , Pain Measurement , Pain Threshold/drug effects , Ruthenium Red/therapeutic use
12.
Yakugaku Zasshi ; 128(11): 1699-705, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18981706

ABSTRACT

The present study was designed to investigate a possible role of vanilloid receptors, CGRP and spleen in the induction of diabetes induced hyperalgesia in mice. Tail flick latency, an index of hyperalgesia, was assessed using analgesiometer. Serum nitrite levels and an index of nitric oxide were analyzed using Griess reaction. Mice were rendered diabetic with streptozotocin (200 mg/kg(-1), i.p.) and kept for 30 days for development of diabetic pain. To explore the involvement of spleen in diabetic pain, spleen homogenate supernatant (SHS) was prepared from spleen of 30th day diabetic mice and administered in normal mice for 14 days. Both in diabetic and SHS treated mice, significant degree of hyperalgesia was developed, suggesting the possible role of spleen derived factor in induction of diabetic pain. Moreover, the levels of nitric oxide were also elevated in 30 day diabetic mice and SHS treated mice. Administration of ruthenium red (1 mg/kg(-1), i.p.), vanilloid receptor antagonist, and sumatriptan (50 mg/kg(-1), i.p.), a CGRP release inhibitor, attenuated diabetes and SHS induced decrease in nociceptive threshold and increase in serum nitrite oxide levels. These results suggest that spleen derived factor induced activation of vanilloid receptors and CGRP release may be contributing in the development of hyperalgesia in diabetic mice.


Subject(s)
Calcitonin Gene-Related Peptide/physiology , Diabetes Mellitus, Experimental/complications , Hyperalgesia/etiology , Spleen/physiology , TRPV Cation Channels/physiology , Animals , Calcitonin Gene-Related Peptide/antagonists & inhibitors , Diabetes Mellitus, Experimental/drug therapy , Female , Hyperalgesia/drug therapy , Male , Mice , Nitric Oxide/metabolism , Ruthenium Red/pharmacology , Ruthenium Red/therapeutic use , Streptozocin , Sumatriptan/pharmacology , Sumatriptan/therapeutic use , TRPV Cation Channels/antagonists & inhibitors
13.
Neuroscience ; 152(2): 511-20, 2008 Mar 18.
Article in English | MEDLINE | ID: mdl-18272293

ABSTRACT

Ankyrin-repeat transient receptor potential 1 (TRPA1) is a member of the transient receptor potential (TRP) channel family and it is found in sensory neurons. In the present study, we found that TRPA1 receptor activation with allyl isothiocyanate or cinnamaldehyde caused dose-dependent spontaneous nociception when injected into the mouse hind paw. Very similar results were obtained when stimulating transient receptor potential vanilloid 1 (TRPV1) receptors with capsaicin. Pretreatment with the TRP receptor antagonist Ruthenium Red (1 nmol/paw) inhibited capsaicin-(0.1 nmol/paw) and allyl isothiocyanate-(1 nmol/paw) induced nociceptive responses. However, the nonselective TRPV1 receptor antagonist capsazepine (1 nmol/paw) and the selective TRPV1 receptor antagonist SB 366791 (1 nmol/paw) only attenuated capsaicin-induced nociception. In contrast, the intrathecal treatment with TRPA1 antisense oligodeoxynucleotide (2.5 nmol/site) and the degeneration of the subset of primary afferent fibers sensitive to capsaicin significantly reduced allyl isothiocyanate-induced nociception. Consequently to TRPA1 antisense oligodeoxynucleotide treatment there was a marked decrease of the expression of TRPA1 receptor in both sciatic nervous and spinal cord segments. Moreover, capsaicin and allyl isothiocyanate-induced nociception were not significantly changed by chemical sympathectomy produced by guanethidine. The previous degranulation of mast cells by compound 48/80 and treatment with antagonist H(1) receptor antagonist pyrilamine (400 microg/paw) both significantly inhibited the capsaicin- and allyl isothiocyanate-induced nociception. The selective NK(1) receptor antagonist N(2)-[(4R)-4-hydroxy-1-(1-methyl-1H-indol-3-yl) carbony-1-L-prolyl]-N-methyl-N-phenylmethyl-3-2-(2-naphtyl)-L-alaninamide (10 nmol/paw) reduced either capsaicin- or allyl isothiocyanate-induced nociception. Collectively, the present findings demonstrate that the TRPA1 agonist allyl isothiocyanate produces a consistent nociceptive response when injected into the mouse paw, an effect that seems to be mediated via activation of TRPA1 receptor and dependent on the capsaicin-sensitive fibers, release of histamine by mast cells and participation of tachykinins. Thus, the TRPA1 receptor has an apparently relevant role in nociceptive processes and the selective TRPA1 antagonist might possess a potential antinociceptive property.


Subject(s)
Capsaicin , Isothiocyanates , Pain/chemically induced , Pain/metabolism , Transient Receptor Potential Channels/metabolism , Analgesics/therapeutic use , Anilides/administration & dosage , Animals , Behavior, Animal , Capsaicin/administration & dosage , Capsaicin/analogs & derivatives , Cinnamates/administration & dosage , Dipeptides/therapeutic use , Dose-Response Relationship, Drug , Drug Interactions , Indoles/therapeutic use , Male , Mice , Pain/prevention & control , Pain Measurement , Ruthenium Red/therapeutic use , TRPA1 Cation Channel , Time Factors , Transient Receptor Potential Channels/agonists , Transient Receptor Potential Channels/antagonists & inhibitors
14.
Int J Oncol ; 17(2): 353-65, 2000 Aug.
Article in English | MEDLINE | ID: mdl-10891547

ABSTRACT

Heavy metals have often been represented, as an uncertain entity related to renal and other risks of toxicity. In favour of this thought there are several lines of evidence, first of all traffic pollution, other evidence that metals such as arsenite, mercury, cadmium or even iron or radioactive heavy metals, that may be introduced into the body by accident, have been responsible of well known pathologies (for example saturnism with lead) or acute toxicity. Therefore, the biological and medical literature have debated on this subject, mainly from the toxicological point of view, rather than studying possible advantages that might come from compounds based on these metals. Exceptions are represented by studies on the role of metal ions in the biochemistry of enzymes and energy production and, although with less emphasis, on their possible use for correcting metabolic malfunctions. Ruthenium, as a metal, has received an even poorer interest and besides the use in histology, neither ruthenium ions nor ruthenium compounds have a clear place in medicine and biology. Nevertheless, since the middle seventies, many studies have been published, showing in a convincible and repetitive manner, the possible advantages of ruthenium as a base for new competitive drugs. The aim of this review is therefore that of critically examining the past and the actual work on ruthenium compounds with emphasis on their proposed role in cancer therapy.


Subject(s)
Antineoplastic Agents/therapeutic use , Carcinoma/drug therapy , Indicators and Reagents/therapeutic use , Neoplasms, Experimental/drug therapy , Ruthenium Compounds/therapeutic use , Animals , Antineoplastic Agents/chemistry , Dimethyl Sulfoxide/chemistry , Dimethyl Sulfoxide/therapeutic use , Drug Screening Assays, Antitumor , Humans , Indicators and Reagents/chemistry , Ruthenium Compounds/chemistry , Ruthenium Red/chemistry , Ruthenium Red/therapeutic use , Solvents/chemistry , Solvents/therapeutic use
15.
Br J Pharmacol ; 114(3): 570-7, 1995 Feb.
Article in English | MEDLINE | ID: mdl-7537589

ABSTRACT

1. Oedema formation induced by intradermal capsaicin has been studied in rabbit skin. The effect of the anti-inflammatory steroid dexamethasone and also of a range of known inhibitors of oedema formation have been investigated in order to elucidate mechanisms involved in capsaicin-induced oedema formation. 2. Oedema formation, in response to intradermally-injected test agents, was measured by the local extravascular accumulation of intravenously injected 125I-labelled albumin. In separate experiments skin blood flow was assessed by the clearance of intradermally-injected 133xenon. 3. Oedema formation induced by intradermal histamine (3 nmol) and bradykinin (1 nmol), when in the presence of vasodilator doses of calcitonin gene-related peptide (CGRP) (3 pmol) or prostaglandin E1, (PGE1) (10 pmol), was significantly inhibited (P < 0.01) in rabbits pretreated with intravenous dexamethasone (3 mg kg-1, -4 h). In contrast dexamethasone had no effect on capsaicin (3 mumol)-induced oedema formation or, on capsaicin (30-100 nmol)-induced blood flow. 4. Oedema formation observed in response to intradermal capsaicin (3 mumol) was significantly inhibited (P < 0.01) when the selective capsaicin antagonist, ruthenium red (3 nmol) was co-injected. This suggests that the mechanism of capsaicin-induced oedema involves activation of sensory nerves. However, oedema was not inhibited when capsaicin was co-injected with the neurokinin NK1 receptor antagonist, RP67580 (10 nmol), the NK2 antagonist SR48960 (10 nmol) or the CGRP antagonist CGRP8-37 (300 pmol). 5. Oedema formation induced by capsaicin was not inhibited when co-injected with the histamine HI receptor antagonist, mepyramine (3 nmol), the PAF antagonist, WEB 2086 (100 nmol), the bradykinin B2 receptor antagonist, Hoel4O (1 nmol), or the cyclo-oxygenase inhibitor, indomethacin (10 nmol),suggesting that these mediators do not play a major role in the capsaicin-induced response.6. Histological analysis of capsaicin-treated skin sites revealed undamaged, intact microvessels and lack of haemorrhage. Further, co-injection of capsaicin with the hydrogen peroxide remover, catalase(2,200 u), had no effect on oedema formation. This suggests that capsaicin does not induce oedema formation secondary to free radical-induced damage.7. These results indicate that capsaicin-induced oedema in rabbit skin involves activation of sensory nerves. However, the oedema is not inhibited by pretreatment with the anti-inflammatory steroid,dexamethasone. Further the mechanisms which lead to the oedema formation observed after intradermal capsaicin remain unknown.


Subject(s)
Capsaicin/toxicity , Edema/chemically induced , Skin/drug effects , Alprostadil/administration & dosage , Alprostadil/pharmacology , Alprostadil/therapeutic use , Animals , Anti-Inflammatory Agents, Non-Steroidal/administration & dosage , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Benzamides/administration & dosage , Benzamides/pharmacology , Benzamides/therapeutic use , Bradykinin/administration & dosage , Bradykinin/toxicity , Calcitonin Gene-Related Peptide/administration & dosage , Calcitonin Gene-Related Peptide/antagonists & inhibitors , Calcitonin Gene-Related Peptide/pharmacology , Calcitonin Gene-Related Peptide/therapeutic use , Capillary Permeability/drug effects , Capsaicin/administration & dosage , Catalase/administration & dosage , Catalase/pharmacology , Catalase/therapeutic use , Dexamethasone/administration & dosage , Dexamethasone/pharmacology , Dexamethasone/therapeutic use , Drug Interactions , Edema/drug therapy , Edema/physiopathology , Histamine/administration & dosage , Histamine/toxicity , Indoles/administration & dosage , Indoles/pharmacology , Indoles/therapeutic use , Indomethacin/administration & dosage , Indomethacin/pharmacology , Indomethacin/therapeutic use , Injections, Intradermal , Injections, Intravenous , Isoindoles , Male , Piperidines/administration & dosage , Piperidines/pharmacology , Piperidines/therapeutic use , Rabbits , Regional Blood Flow/drug effects , Ruthenium Red/administration & dosage , Ruthenium Red/pharmacology , Ruthenium Red/therapeutic use , Skin/blood supply , Skin/pathology , Substance P/antagonists & inhibitors
16.
Cardiovasc Res ; 25(4): 337-42, 1991 Apr.
Article in English | MEDLINE | ID: mdl-1715813

ABSTRACT

STUDY OBJECTIVE: The aim was to investigate the effect of attenuating mitochondrial calcium uptake with ruthenium red on myocardial function and the resultant necrosis following prolonged ischaemia and reperfusion in isolated rat hearts. Mitochondrial dysfunction, secondary to increased calcium uptake, has been implicated as an important mediator of reperfusion injury in the heart. DESIGN: To examine the role of mitochondrial calcium uptake in mediating ischaemic and reperfusion injury, isolated rat hearts were perfused with ruthenium red (n = 6), a polysaccharide dye which inhibits calcium uptake by mitochondria, and were compared to control perfused hearts (n = 7). After stabilisation, hearts were subjected to 60 min no flow ischaemia, immediately followed by 40 min reperfusion. EXPERIMENTAL MATERIAL: Hearts were used from male Wistar rats weighing 300-350 g. MEASUREMENTS AND MAIN RESULTS: Cardiac high energy phosphates (ATP, phosphocreatine, inorganic phosphate) and pH were continuously monitored during ischaemia and reperfusion using phosphorus magnetic resonance spectroscopy. Contractility (dP/dT), coronary flow, creatine kinase release, and the time to the onset of ischaemic contracture were also measured. No differences in metabolic abnormalities or time to peak contraction during ischaemia were found between groups, suggesting that ruthenium red does not alter the metabolic consequences of ischaemia. However, upon reperfusion, the following differences in the ruthenium red perfused hearts were observed when compared to control hearts (p less than 0.05): ATP and phosphocreatine recovery were more complete, myocardial contractility was greater, coronary flow was greater, and myocyte necrosis was attenuated. CONCLUSIONS: Combined with the known inhibitory effect of ruthenium red on mitochondrial calcium uptake, these data suggest that an important component of myocardial injury following ischaemia and reperfusion in the isolated rat heart is the result of mitochondrial calcium accumulation.


Subject(s)
Myocardial Reperfusion Injury/prevention & control , Ruthenium Red/therapeutic use , Adenosine Triphosphate/metabolism , Animals , Calcium/metabolism , Coronary Circulation/drug effects , Male , Mitochondria, Heart/drug effects , Mitochondria, Heart/metabolism , Myocardial Contraction/drug effects , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/pathology , Myocardium/pathology , Perfusion , Phosphocreatine/metabolism , Rats , Rats, Inbred Strains
17.
Gan ; 71(1): 151-4, 1980 Feb.
Article in English | MEDLINE | ID: mdl-6155302

ABSTRACT

The growth of subcutaneously transplanted Lewis lung carcinoma in BDF1 mice was inhibited by an inorganic dye, Ruthenium Red. The mean volume of the tumors of the mice which were given Ruthenium Red by daily ip injection of 5 mg/kg on days 1 approximately 10 was 28 and 39% of that of the controls at days 18 and 23 after transplantation, respectively. The median survival time (MST) of the treated mice was 27.8 days while that of the control was 23.0 days. The higher MST (32.5 days) was obtained when the dye was given at 2.5 mg/kg for 1 approximately 20 days, although the tumor volume in this group of mice was 51% of that of the control at day 18.


Subject(s)
Antineoplastic Agents/therapeutic use , Lung Neoplasms/drug therapy , Ruthenium Red/therapeutic use , Ruthenium/therapeutic use , Animals , Male , Mice , Neoplasms, Experimental/drug therapy , Rats , Ruthenium Red/administration & dosage , Ruthenium Red/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...