Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 127
Filter
1.
J Pharmacol Sci ; 152(3): 167-177, 2023 Jul.
Article in English | MEDLINE | ID: mdl-37257944

ABSTRACT

Cisplatin, a platinum-based anticancer drug used frequently in cancer treatment, causes skeletal muscle atrophy. It was predicted that the proteolytic pathway is enhanced as the mechanism of this atrophy. Therefore, we investigated whether a platinum-based anticancer drug affects the expression of the major proteins of skeletal muscle, myosin heavy chain (MyHC). Mice were injected with cisplatin or oxaliplatin for four consecutive days. C2C12 myotubes were treated using cisplatin and oxaliplatin. Administration of platinum-based anticancer drug reduced quadriceps mass and muscle strength compared to the control group. Protein levels of all MyHC isoforms were reduced in the platinum-based anticancer drug groups. However, only Myh2 (MyHC-IIa) gene expression in skeletal muscle of mice treated with platinum-based anticancer drugs was found to be reduced. Treatment of C2C12 myotubes with platinum-based anticancer drugs reduced the protein levels of all MyHCs, and treatment with the proteasome inhibitor MG-132 restored this reduction. The expression of Mef2c, which was predicted to act upstream of Myh2, was reduced in the skeletal muscle of mice treated systemically with platinum-based anticancer drug. Degradation of skeletal muscle MyHCs by proteasomes may be a factor that plays an important role in muscle mass loss in platinum-based anticancer drug-induced muscle atrophy.


Subject(s)
Antineoplastic Agents , Myosin Heavy Chains , Mice , Animals , Myosin Heavy Chains/genetics , Myosin Heavy Chains/metabolism , Down-Regulation , Cisplatin , Platinum/metabolism , Oxaliplatin , Muscle, Skeletal/metabolism , Muscle Fibers, Skeletal/metabolism , Antineoplastic Agents/pharmacology , Protein Isoforms/genetics , Protein Isoforms/metabolism , Atrophy/metabolism
2.
Chem Pharm Bull (Tokyo) ; 71(4): 289-298, 2023 Apr 01.
Article in English | MEDLINE | ID: mdl-36709972

ABSTRACT

Oral disulfiram (DSF) has been used clinically for alcohol dependence and recently has been found to have antitumor activity. A transdermal delivery system would be useful for maintaining drug concentration and reducing the frequency of administration of DSF for cancer treatment. Penetrating the stratum corneum (SC) barrier is a challenge to the transdermal delivery of DSF. Therefore, we investigated the promoting effects and mechanism of action of the combination of oleic acid (OA) and Tween 80 on the skin permeation of DSF. Hairless mouse skin was exposed to OA and Tween 80, combined in various ratios (1 : 0, 2 : 1, 1 : 1, 1 : 2, and 0 : 1). A permeation experiment was performed, and total internal reflection IR spectroscopic measurements, differential scanning calorimetry, and synchrotron radiation X-ray diffraction measurements were taken of the SC with each applied formulation. The combination of OA and Tween 80 further enhanced the absorption-promoting effect of DSF, compared with individual application. The peak of the CH2 inverse symmetric stretching vibration near the skin surface temperature was shifted by a high frequency due to the application of OA, and DSF solubility increased in response to Tween 80. We believe that the increased fluidity of the intercellular lipids due to OA and the increased solubility of DSF due to Tween 80 promoted the absorption of DSF. Our study clarifies the detailed mechanism of action of the skin permeation and promoting effect of DSF through the combined use of OA and Tween 80, contributing to the development of a transdermal preparation of DSF.


Subject(s)
Oleic Acid , Polysorbates , Mice , Animals , Oleic Acid/analysis , Oleic Acid/chemistry , Oleic Acid/pharmacology , Polysorbates/analysis , Polysorbates/pharmacology , Disulfiram/pharmacology , Disulfiram/analysis , Skin , Administration, Cutaneous
3.
Int J Mol Sci ; 24(21)2023 Oct 31.
Article in English | MEDLINE | ID: mdl-37958806

ABSTRACT

The aim of this research was to test the efficacy and potential clinical application of intranasal administration of galanin-like peptide (GALP) as an anti-obesity treatment under the hypothesis that GALP prevents obesity in mice fed a high-fat diet (HFD). Focusing on the mechanism of regulation of lipid metabolism in peripheral tissues via the autonomic nervous system, we confirmed that, compared with a control (saline), intranasally administered GALP prevented further body weight gain in diet-induced obesity (DIO) mice with continued access to an HFD. Using an omics-based approach, we identified several genes and metabolites in the liver tissue of DIO mice that were altered by the administration of intranasal GALP. We used whole-genome DNA microarray and metabolomics analyses to determine the anti-obesity effects of intranasal GALP in DIO mice fed an HFD. Transcriptomic profiling revealed the upregulation of flavin-containing dimethylaniline monooxygenase 3 (Fmo3), metallothionein 1 and 2 (Mt1 and Mt2, respectively), and the Aldh1a3, Defa3, and Defa20 genes. Analysis using the DAVID tool showed that intranasal GALP enhanced gene expression related to fatty acid elongation and unsaturated fatty acid synthesis and downregulated gene expression related to lipid and cholesterol synthesis, fat absorption, bile uptake, and excretion. Metabolite analysis revealed increased levels of coenzyme Q10 and oleoylethanolamide in the liver tissue, increased levels of deoxycholic acid (DCA) and taurocholic acid (TCA) in the bile acids, increased levels of taurochenodeoxycholic acid (TCDCA), and decreased levels of ursodeoxycholic acid (UDCA). In conclusion, intranasal GALP administration alleviated weight gain in obese mice fed an HFD via mechanisms involving antioxidant, anti-inflammatory, and fatty acid metabolism effects and genetic alterations. The gene expression data are publicly available at NCBI GSE243376.


Subject(s)
Diet, High-Fat , Galanin-Like Peptide , Mice , Animals , Diet, High-Fat/adverse effects , Galanin-Like Peptide/metabolism , Galanin-Like Peptide/pharmacology , Oligonucleotide Array Sequence Analysis , Transcriptome , Administration, Intranasal , Obesity/etiology , Obesity/genetics , Liver/metabolism , Weight Gain , Metabolome , Lipid Metabolism , Fatty Acids/metabolism , Mice, Inbred C57BL
4.
Biol Pharm Bull ; 45(8): 1208-1212, 2022.
Article in English | MEDLINE | ID: mdl-35908904

ABSTRACT

We have previously reported that swellings caused by haptens, such as 2,4,6-trinitrochlorobenzene (TNCB), may be associated with the extracellular signal-regulated kinase (ERK)-induced proliferation pathway. However, the involvement of the Spred/Sprouty family as critical negative regulators of the Ras/Raf/ERK signaling pathway at disease sites is not well-established. Thus, in the present study, the effects of hapten-challenge on the expression levels of genes and proteins associated with the Spred/Sprouty family in the ear of mice were investigated. The activation of ERK and epidermal growth factor receptor (EGFR) tyrosine kinase was inhibited by their selective inhibitors, namely, U0126 and PD168393, respectively. Twenty-four hours after the final challenge by the haptens TNCB, 2,4-dinitrofluorobenzene, or oxazolone, ear thickness was augmented by challenge with all haptens and the gene expression levels of Spred1, Spred2, Sprouty1, and Sprouty2 in swelling induced by all haptens were significantly decreased. Furthermore, Spred2, Sprouty1, and Sprouty2 genes were decreased in the epidermis and dermis of the TNCB-challenged ear. In conclusion, it is possible that the mechanism of hapten-challenge-induced skin thickening involves not only the enhancement of cell proliferative functions via the activation of ERK by EGFR tyrosine kinase activation but also the decreases expression of Spred/Sprouty family members.


Subject(s)
Dermatitis, Contact , Repressor Proteins , Animals , ErbB Receptors/genetics , Extracellular Signal-Regulated MAP Kinases/metabolism , Mice , Picryl Chloride , Protein-Tyrosine Kinases , Repressor Proteins/metabolism
5.
Biol Pharm Bull ; 45(7): 910-918, 2022.
Article in English | MEDLINE | ID: mdl-35786599

ABSTRACT

Cisplatin is a chemotherapy drug used to treat a variety of cancers. Muscle loss in cancer patients is associated with increased cancer-related mortality. Previously, we suggested that cisplatin administration increases the atrophic gene expressions of ubiquitin E3 ligases, such as atrogin-1 and muscle RING finger-1 (MuRF1), which may lead to muscle atrophy. In this study, C57BL/6J mice were treated with cisplatin (3 mg/kg, intraperitoneally) or saline for 4 consecutive days. Twenty-four hours after the final injection of cisplatin, quadriceps muscles were removed from the mice. The gene expression of Psma and Psmb, which comprise the 20S proteasome, was upregulated by cisplatin administration in the quadriceps muscle of mouse. Systemic administration of cisplatin significantly reduced not only the quadriceps muscle mass but also the diameter of the myofibers. In addition, bortezomib (0.125 mg/kg, intraperitoneally) was administered 30 min before each cisplatin treatment. The co-administration of bortezomib, a proteasome inhibitor, significantly recovered the reductions in the mass of quadriceps and myofiber diameter, although it did not recover the decline in the forelimb and forepaw strength induced by cisplatin. Increased 20S proteasome abundance may play a significant role in the development of cisplatin-induced muscle atrophy. During cisplatin-induced skeletal muscle atrophy, different mechanisms may be involved between loss of muscle mass and strength. In addition, it is suggested that bortezomib has essentially no effect on cisplatin-induced muscle atrophy.


Subject(s)
Cisplatin , Proteasome Endopeptidase Complex , Animals , Bortezomib , Mice , Mice, Inbred C57BL , Muscular Atrophy/chemically induced , Muscular Atrophy/drug therapy
6.
Lung ; 200(5): 591-599, 2022 10.
Article in English | MEDLINE | ID: mdl-35930050

ABSTRACT

PURPOSE: Extracellular acidification is a major component of tissue inflammation, including airway inflammation. The extracellular proton-sensing mechanisms are inherent in various cells including airway structural cells, although their physiological and pathophysiological roles in bronchial smooth muscles (BSMs) are not fully understood. In the present study, to explore the functional role of extracellular acidification on the BSM contraction, the isolated mouse BSMs were exposed to acidic pH under contractile stimulation. METHODS AND RESULTS: The RT-PCR analyses revealed that the proton-sensing G protein-coupled receptors were expressed both in mouse BSMs and cultured human BSM cells. In the mouse BSMs, change in the extracellular pH from 8.0 to 6.8 caused an augmentation of contraction induced by acetylcholine. Interestingly, the acidic pH-induced BSM hyper-contraction was further augmented in the mice that were sensitized and repeatedly challenged with ovalbumin antigen. In this animal model of asthma, upregulations of G protein-coupled receptor 68 (GPR68) and GPR65, that were believed to be coupled with Gq and Gs proteins respectively, were observed, indicating that the acidic pH could cause hyper-contraction probably via an activation of GPR68. However, psychosine, a putative antagonist for GPR68, failed to block the acidic pH-induced responses. CONCLUSION: These findings suggest that extracellular acidification contributes to the airway hyperresponsiveness, a characteristic feature of bronchial asthma. Further studies are required to identify the receptor(s) responsible for sensing extracellular protons in BSM cells.


Subject(s)
Asthma , Bronchial Hyperreactivity , Acetylcholine/adverse effects , Acetylcholine/metabolism , Animals , Bronchi , Bronchial Hyperreactivity/metabolism , Humans , Hydrogen-Ion Concentration , Inflammation/metabolism , Mice , Mice, Inbred BALB C , Muscle, Smooth/metabolism , Ovalbumin , Protons , Psychosine/adverse effects , Psychosine/metabolism , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism
7.
J Card Surg ; 37(8): 2338-2347, 2022 Aug.
Article in English | MEDLINE | ID: mdl-35545926

ABSTRACT

OBJECTIVE: Although hemodilution during hypothermic cardiopulmonary bypass (CPB) had been thought to improve microcirculation and reduce blood viscosity, there has been no report investigating the effect of low nadir hematocrit (Hct) values caused by severe hemodilution on the surgical outcomes of patients with acute type A aortic dissection (ATAAD). METHODS: We retrospectively reviewed 112 consecutive patients who emergently underwent emergency surgical repair of ATAAD at our institution. The patients were classified into the high Hct (nadir Hct ≥ 21% during CPB; n = 51) and low Hct (nadir Hct < 21% during CPB; n = 61) groups. After propensity score matching of preoperative characteristics, surgical outcomes were compared between the groups. RESULTS: Although there was no difference in the surgical procedure, longer CPB time and more blood transfusion during surgery were needed in the low Hct group than in the high Hct group. After surgery, estimated glomerular filtration rate was significantly lower (p = .015), lactaic acid was higher (p = .045), and intubation time was longer (p = .018) in the low Hct group than in the high Hct group, although there was no difference in hospital mortality between the groups. The AUC of the nadir Hct during CPB as a prognostic indicator of prolonged postoperative ventilator support was 0.8, with the highest accuracy at 16.7% (sensitivity 88%, specificity 76.9%). In all cohorts, female sex was an independent risk factor for a lower nadir Hct value of <21% during CPB. CONCLUSION: A lower nadir Hct value of <21% during CPB may be associated with postoperative renal dysfunction and prolonged ventilator support in patients with ATAAD.


Subject(s)
Aortic Dissection , Cardiopulmonary Bypass , Aortic Dissection/etiology , Aortic Dissection/surgery , Cardiopulmonary Bypass/adverse effects , Female , Hematocrit , Hemodilution , Humans , Retrospective Studies
8.
Am J Physiol Lung Cell Mol Physiol ; 319(5): L786-L793, 2020 11 01.
Article in English | MEDLINE | ID: mdl-32877227

ABSTRACT

Bronchomotor tone is regulated by contraction and relaxation of airway smooth muscle (ASM). A weakened ASM relaxation might be a cause of airway hyperresponsiveness (AHR), a characteristic feature of bronchial asthma. Pituitary adenylyl cyclase-activating polypeptide (PACAP) is known as a mediator that causes ASM relaxation. To date, whether or not the PACAP responsiveness is changed in asthmatic ASM is unknown. The current study examined the hypothesis that relaxation induced by PACAP is reduced in bronchial smooth muscle (BSM) of allergic asthma. The ovalbumin (OA)-sensitized mice were repeatedly challenged with aerosolized OA to induce asthmatic reaction. Twenty-four hours after the last antigen challenge, the main bronchial smooth muscle (BSM) tissues were isolated. Tension study showed a BSM hyperresponsiveness to acetylcholine in the OA-challenged mice. Both quantitative RT-PCR and immunoblot analyses revealed a significant decrease in PAC1 receptor expression in BSMs of the diseased mice. Accordingly, in the antigen-challenged group, the PACAP-induced PAC1 receptor-mediated BSM relaxation was significantly attenuated, whereas the relaxation induced by vasoactive intestinal polypeptide was not changed. These findings suggest that the relaxation induced by PACAP is impaired in BSMs of experimental asthma due to a downregulation of its binding partner PAC1 receptor. Impaired BSM responsiveness to PACAP might contribute to the AHR in asthma.


Subject(s)
Asthma/metabolism , Bronchi/metabolism , Muscle, Smooth/metabolism , Pituitary Adenylate Cyclase-Activating Polypeptide/metabolism , Surface-Active Agents/metabolism , Animals , Bronchial Hyperreactivity/metabolism , Mice , Muscle Relaxation/drug effects , Muscle Relaxation/physiology , Respiratory Hypersensitivity/metabolism , Vasoactive Intestinal Peptide/metabolism
9.
Toxicol Appl Pharmacol ; 403: 115165, 2020 09 15.
Article in English | MEDLINE | ID: mdl-32738330

ABSTRACT

We previously demonstrated that cisplatin administration in mice induces muscle atrophy and an increase in the expression of two muscle-specific ubiquitin E3 ligase genes, muscle ring finger protein 1 (MuRF1), and atrophy gene-1 (atrogin-1), in skeletal muscle. Ubiquitination serves as a degradation signal in both the ubiquitin-proteasome and selective autophagy pathways. In the present study, we investigated changes in the expression of ubiquitin and ubiquitinated proteins and their degradation pathways. Ubiquitin and ubiquitinated protein levels were increased by cisplatin compared with those in the vehicle and dietary restriction (DR) groups. To quantify the levels of ubiquitin and ubiquitinated proteins, we conducted a dot blot assay using an anti-ubiquitin antibody. The expression of ubiquitin was also significantly increased by cisplatin compared with that in the vehicle and DR groups. Since the ubiquitin proteins were upregulated by cisplatin, we measured the mRNA levels of the ubiquitin genes: Ubb, Ubc, Rps27a, and Uba52. All these four genes were increased by cisplatin administration compared with those in both the vehicle-treated and DR groups in quadriceps muscle tissue. The anti-ubiquitin antibody-sensitive bands increased when C2C12 myotubes were treated with cisplatin. Furthermore, MG-132 (26 s proteasome inhibitor), but not bafilomycin A1 (autophagy inhibitor), caused a further increase in expression. In conclusion, ubiquitin and ubiquitinated proteins are upregulated in cisplatin-induced muscle atrophy. Cisplatin-induced ubiquitinated proteins are degraded by the 26 s proteasome pathway.


Subject(s)
Cisplatin/toxicity , Gene Expression Regulation/drug effects , Muscular Atrophy/chemically induced , Ubiquitinated Proteins/metabolism , Up-Regulation/drug effects , Animals , Antineoplastic Agents/toxicity , Cell Line , Gene Expression Regulation/physiology , Male , Mice , Mice, Inbred C57BL , Myoblasts/drug effects , Ubiquitinated Proteins/genetics
10.
Int J Mol Sci ; 21(21)2020 Oct 27.
Article in English | MEDLINE | ID: mdl-33121100

ABSTRACT

In allergic bronchial asthma, an increased smooth muscle contractility of the airways is one of the causes of the airway hyperresponsiveness (AHR). Increasing evidence also suggests a possible involvement of microRNAs (miRNAs) in airway diseases, including asthma, although their roles in function and pathology largely unknown. The current study aimed to determine the role of a miRNA, miR-140-3p, in the control of protein expression of CD38, which is believed to regulate the contraction of smooth muscles, including the airways. In bronchial smooth muscles (BSMs) of the mice that were actively sensitized and repeatedly challenged with ovalbumin antigen, an upregulation of CD38 protein concurrently with a significant reduction of miR-140-3p was observed. In cultured human BSM cells (hBSMCs), transfection with a synthetic miR-140-3p inhibitor caused an increase in CD38 protein, indicating that its basal protein expression is regulated by endogenous miR-140-3p. Treatment of the hBSMCs with interleukin-13 (IL-13), an asthma-related cytokine, caused both an upregulation of CD38 protein and a downregulation of miR-140-3p. Transfection of the hBSMCs with miR-140-3p mimic inhibited the CD38 protein upregulation induced by IL-13. On the other hand, neither a CD38 product cyclic ADP-ribose (cADPR) nor its antagonist 8-bromo-cADPR had an effect on the BSM contraction even in the antigen-challenged mice. Taken together, the current findings suggest that the downregulation of miR-140-3p induced by IL-13 might cause an upregulation of CD38 protein in BSM cells of the disease, although functional and pathological roles of the upregulated CD38 are still unclear.


Subject(s)
ADP-ribosyl Cyclase 1/genetics , Asthma/genetics , Bronchi/metabolism , Hypersensitivity/complications , Membrane Glycoproteins/genetics , MicroRNAs/genetics , ADP-ribosyl Cyclase 1/metabolism , Animals , Asthma/etiology , Asthma/metabolism , Bronchi/cytology , Cell Line , Disease Models, Animal , Gene Expression Regulation/drug effects , Humans , Hypersensitivity/genetics , Hypersensitivity/metabolism , Interleukin-13/pharmacology , Male , Membrane Glycoproteins/metabolism , Mice , Mice, Inbred BALB C , Myocytes, Smooth Muscle/metabolism , Ovalbumin/adverse effects
11.
J Pharmacol Sci ; 141(2): 106-110, 2019 Oct.
Article in English | MEDLINE | ID: mdl-31679962

ABSTRACT

It has recently been exhibited that Rac1 expression is increased in the bronchial tissue of a murine model with repeated antigen-challenged airway hyperresponsiveness (AHR). In the present study, the role of Rac1 in endothelin-1 (ET-1)-induced bronchial contraction and myosin light chain (MLC) phosphorylation was examined in AHR mice. Enhanced reactions in AHR mice were prevented by the Rac1 inhibitor NSC23766. These findings suggest that increased activation of Rac1 might be responsible for the enhancement of the bronchial contraction induced by ET-1 in AHR.


Subject(s)
Bronchial Hyperreactivity/metabolism , Bronchoconstriction , Endothelin-1/metabolism , rac1 GTP-Binding Protein/metabolism , Aminoquinolines/metabolism , Aminoquinolines/pharmacology , Animals , Bronchi/metabolism , Disease Models, Animal , Male , Mice , Mice, Inbred BALB C , Muscle, Smooth/metabolism , Muscle, Smooth/physiopathology , Myosin Light Chains/chemistry , Myosin Light Chains/metabolism , Phosphorylation , Pyrimidines/metabolism , Pyrimidines/pharmacology
12.
Biol Pharm Bull ; 42(9): 1605-1607, 2019 Sep 01.
Article in English | MEDLINE | ID: mdl-31243195

ABSTRACT

Recently, we demonstrated that Rac1 upregulation is involved in augmented bronchial smooth muscle (BSM) contractions of antigen-challenged mice. However, change in G protein-coupled receptor (GPCR)-induced Rac1 activation remains unknown in BSMs of repeatedly antigen-challenged (Chal.) mice. We here examined carbachol (CCh)-induced Rac1 activation in BSMs of Chal. mice. Gene expression levels of both Rac1 and Rac-guanine nucleotide exchange factors (GEFs), such as Tiam1 and Trio, were increased in BSMs of Chal. mice. Furthermore, CCh-induced Rac1 activation was inhibited by pretreatment with Rac1-GEF inhibitor NSC23766 and Rac1 inhibitor EHT1864 in BSMs of sensitized-control (S.C.) and Chal. mice. Compared with S.C. mice, CCh-induced Rac1 activation was increased in BSMs of Chal. mice. In conclusion, we reported that increased CCh-induced Rac1 activation via Tiam1 and Trio upregulation, in addition to upregulate Rac1, may be involved in increased CCh-induced BSM contractions in Chal. mice.


Subject(s)
Bronchi/physiology , Guanine Nucleotide Exchange Factors/physiology , Muscle Contraction/physiology , Muscle, Smooth/physiology , Neuropeptides/physiology , Phosphoproteins/physiology , Protein Serine-Threonine Kinases/physiology , T-Lymphoma Invasion and Metastasis-inducing Protein 1/physiology , rac1 GTP-Binding Protein/physiology , Aminoquinolines/pharmacology , Animals , Antigens , Asthma/genetics , Asthma/physiopathology , Bronchi/drug effects , Carbachol , Guanine Nucleotide Exchange Factors/genetics , Male , Mice, Inbred BALB C , Muscarinic Agonists , Muscle Contraction/drug effects , Muscle, Smooth/drug effects , Neuropeptides/antagonists & inhibitors , Neuropeptides/genetics , Ovalbumin , Phosphoproteins/genetics , Protein Serine-Threonine Kinases/genetics , Pyrimidines/pharmacology , Pyrones/pharmacology , Quinolines/pharmacology , T-Lymphoma Invasion and Metastasis-inducing Protein 1/genetics , Up-Regulation , rac1 GTP-Binding Protein/antagonists & inhibitors , rac1 GTP-Binding Protein/genetics
13.
Clin Exp Pharmacol Physiol ; 46(1): 19-28, 2019 01.
Article in English | MEDLINE | ID: mdl-30137654

ABSTRACT

Dexamethasone for antiemetic therapy is typically administered with anticancer drugs such as cisplatin. We previously reported that cisplatin upregulates the muscle-specific E3 ubiquitin ligase genes, namely muscle ring-finger protein 1 (MuRF1) and atrophy gene-1 (atrogin-1), and promotes muscle atrophy in mice. It is well known that dexamethasone causes upregulation of MuRF1 and Atrogin-1 expression in skeletal muscles. Although it is speculated that a combination of dexamethasone and cisplatin worsens muscle atrophy, there are no reports based on research. We thereby investigated the effects of cisplatin and dexamethasone, alone or in combination, on the expression of MuRF1 and Atrogin-1 in murine skeletal muscles and C2C12 myotubes. Mice were intraperitoneally injected with cisplatin or the vehicle control once daily for 4 days. Dexamethasone or the vehicle control was subcutaneously administered 30 minutes prior to the administration of cisplatin. Dexamethasone enhanced MuRF1 and Atrogin-1 gene expression upregulated by cisplatin in murine quadriceps muscles and C2C12 myotubes. Cisplatin-caused upregulation of myostatin and downregulation of IGF-1 gene expression were also enhanced by co-administration of dexamethasone in murine quadriceps muscles and C2C12 myotubes. This study shows that the combination treatment of cisplatin and dexamethasone exacerbated muscle atrophy in mice. Therefore, this treatment regimen might exacerbate muscle atrophy in cancer patients.


Subject(s)
Cisplatin/adverse effects , Dexamethasone/adverse effects , Muscular Atrophy/chemically induced , Animals , Body Weight/drug effects , Drug Synergism , Gene Expression Regulation/drug effects , Mice , Muscle Fibers, Skeletal/drug effects , Muscle Fibers, Skeletal/metabolism , Muscle Fibers, Skeletal/pathology , Muscle Proteins/genetics , Muscular Atrophy/genetics , Muscular Atrophy/pathology , SKP Cullin F-Box Protein Ligases/genetics , Tripartite Motif Proteins/genetics , Ubiquitin-Protein Ligases/genetics
14.
Am J Physiol Lung Cell Mol Physiol ; 314(1): L215-L223, 2018 01 01.
Article in English | MEDLINE | ID: mdl-28982738

ABSTRACT

Progranulin (PGRN) is a growth factor with multiple biological functions and has been suggested as an endogenous inhibitor of Tumor necrosis factor-α (TNF-α)-mediated signaling. TNF-α is believed to be one of the important mediators of the pathogenesis of asthma, including airway hyperresponsiveness (AHR). In the present study, effects of recombinant PGRN on TNF-α-mediated signaling and antigen-induced hypercontractility were examined in bronchial smooth muscles (BSMs) both in vitro and in vivo. Cultured human BSM cells (hBSMCs) and male BALB/c mice were used. The mice were sensitized and repeatedly challenged with ovalbumin antigen. Animals also received intranasal administrations of recombinant PGRN into the airways 1 h before each antigen inhalation. In hBSMCs, PGRN inhibited both the degradation of IκB-α (an index of NF-κB activation) and the upregulation of RhoA (a contractile machinery-associated protein that contributes to the BSM hyperresponsiveness) induced by TNF-α, indicating that PGRN has an ability to inhibit TNF-α-mediated signaling also in the BSM cells. In BSMs of the repeatedly antigen-challenged mice, an augmented contractile responsiveness to acetylcholine with an upregulation of RhoA was observed: both the events were ameliorated by pretreatments with PGRN intranasally. Interestingly, a significant decrease in PGRN expression was found in the airways of the repeatedly antigen-challenged mice rather than those of control animals. In conclusion, exogenously applied PGRN into the airways ameliorated the antigen-induced BSM hyperresponsiveness, probably by blocking TNF-α-mediated response. Increasing PGRN levels might be a promising therapeutic for AHR in allergic asthma.


Subject(s)
Asthma/physiopathology , Bronchi/physiopathology , Bronchial Hyperreactivity/prevention & control , Intercellular Signaling Peptides and Proteins/administration & dosage , Muscle, Smooth/pathology , Recombinant Proteins/administration & dosage , Respiratory Hypersensitivity/prevention & control , Administration, Intranasal , Animals , Bronchial Hyperreactivity/etiology , Bronchial Hyperreactivity/metabolism , Cells, Cultured , Granulins , Humans , Male , Mice , Mice, Inbred BALB C , Muscle, Smooth/metabolism , Progranulins , Respiratory Hypersensitivity/etiology , Respiratory Hypersensitivity/metabolism , Signal Transduction
15.
Clin Exp Pharmacol Physiol ; 45(1): 27-33, 2018 Jan.
Article in English | MEDLINE | ID: mdl-28762515

ABSTRACT

Contact dermatitis is a form of delayed-type hypersensitivity characterized by localized thickening, papules, redness and vesicles of the skin. A model of contact dermatitis involving repeated challenge of a hapten is adapted to assess dermatitis as characterized by skin thickening. Recently, it was reported that neutrophils have crucial roles in contact hypersensitivity. We thus examined the involvement of CXC chemokines bearing the glutamic acid-leucine-arginine (ELR) motif ("ELR+ chemokines") and neutrophils in the ear swelling induced by 2,4,6-trinitrochlorobenzene (TNCB) challenges in the present study. Mice were sensitized by application of TNCB on their abdominal skin. They were then challenged thrice with TNCB to the ear. The CXCR2 antagonist SB225002 (9 mg/kg, i.p.) was administered before each TNCB challenge. Gene expressions and protein levels of the ELR+ chemokines CXCL1, 2 and 5 was increased markedly in mouse ear after the final TNCB challenge. In addition, we indicated that gene expression of CXCL1 was enhanced in the epidermis and dermis upon TNCB challenge. Expression of the CXCL2 gene was enhanced in the epidermis, and that of the CXCL5 gene was enhanced in the dermis. The swelling induced by TNCB challenges was significantly attenuated by SB225002. Furthermore, the increases in myeloperoxidase activity, and expression of myeloperoxidase and neutrophil elastase induced by TNCB challenge in mouse ear were inhibited by SB225002. These data suggest that ear swelling resulting from TNCB challenges might be concerned by upregulated ELR+ chemokine-induced neutrophil recruitment.


Subject(s)
Chemokines, CXC/chemistry , Chemokines, CXC/metabolism , Dermatitis, Contact/immunology , Dermatitis, Contact/metabolism , Neutrophil Infiltration/drug effects , Picryl Chloride/adverse effects , Amino Acid Motifs , Animals , Dermatitis, Contact/etiology , Female , Mice, Inbred BALB C , Receptors, Interleukin-8B/antagonists & inhibitors
16.
Int J Mol Sci ; 19(10)2018 Oct 05.
Article in English | MEDLINE | ID: mdl-30301147

ABSTRACT

Prostaglandin D2 (PGD2) is one of the key lipid mediators of allergic airway inflammation, including bronchial asthma. However, the role of PGD2 in the pathogenesis of asthma is not fully understood. In the present study, the effect of PGD2 on smooth muscle contractility of the airways was determined to elucidate its role in the development of airway hyperresponsiveness (AHR). In isolated bronchial smooth muscles (BSMs) of naive mice, application of PGD2 (10-9⁻10-5 M) had no effect on the baseline tension. However, when the tissues were precontracted partially with 30 mM K⁺ (in the presence of 10-6 M atropine), PGD2 markedly augmented the contraction induced by the high K⁺ depolarization. The PGD2-induced augmentation of contraction was significantly inhibited both by 10-6 M laropiprant (a selective DP1 antagonist) and 10-7 M Y-27632 (a Rho-kinase inhibitor), indicating that a DP1 receptor-mediated activation of Rho-kinase is involved in the PGD2-induced BSM hyperresponsiveness. Indeed, the GTP-RhoA pull-down assay revealed an increase in active form of RhoA in the PGD2-treated mouse BSMs. On the other hand, in the high K⁺-depolarized cultured human BSM cells, PGD2 caused no further increase in cytosolic Ca2+ concentration. These findings suggest that PGD2 causes RhoA/Rho-kinase-mediated Ca2+ sensitization of BSM contraction to augment its contractility. Increased PGD2 level in the airways might be a cause of the AHR in asthma.


Subject(s)
Bronchi/metabolism , Calcium/metabolism , Cytosol/metabolism , Muscle Contraction/drug effects , Muscle, Smooth/drug effects , Prostaglandin D2/pharmacology , Animals , Atropine/pharmacology , Bronchial Hyperreactivity/metabolism , Humans , Indoles/pharmacology , Male , Mice , Mice, Inbred BALB C , Potassium/pharmacology , Primary Cell Culture , Receptors, Prostaglandin/drug effects
17.
Pflugers Arch ; 469(11): 1495-1505, 2017 11.
Article in English | MEDLINE | ID: mdl-28762162

ABSTRACT

Cisplatin, a platinum-based anti-cancer drug, is one of the most effective broad-spectrum anti-cancer agents used against various cancers. It has been recently suggested that low skeletal muscle mass is predictive of mortality in patients with cancer. Although several molecules produced by the actual tumor itself contribute to skeletal muscle impairment, we recently suggested that the administration of cisplatin could increase levels of muscle RING finger-1 (MuRF1) and atrogin-1, possibly leading to muscle atrophy in the mouse. Exercise is an important factor that induces muscle protein synthesis and muscle hypertrophy by enhancing the positive effects of the Akt/mTOR/p70S6 kinase pathway. In the present study, we therefore investigated the effect of treadmill exercise on cisplatin-induced muscle atrophy. C57BL/6J mice were treated with cisplatin (3 mg/kg, i.p.) or saline for four consecutive days. On day 4, the quadriceps and gastrocnemius muscles were isolated from the mice. The animals in the treadmill exercise groups were forced to run on a motorized treadmill for 20 min once a day for 9 days. In addition to muscle mass, the decrease in myofiber diameter associated with cisplatin administration was significantly restored by treadmill exercise. This exercise also significantly attenuated cisplatin-induced upregulation of MuRF1 and atrogin-1 in quadriceps and gastrocnemius muscle. The decreased Akt, p70S6 kinase, and Foxo3a phosphorylation observed with cisplatin treatment was significantly recovered by treadmill exercise in both the muscles. In the present study, myostatin (Mstn) gene expression, upregulated by cisplatin administration, was also attenuated by treadmill exercise. These findings suggest that treadmill exercise could attenuate cisplatin-induced muscle atrophy, at least partially, and could improve prognosis.


Subject(s)
Cisplatin/pharmacology , Muscle, Skeletal/physiology , Muscular Atrophy/chemically induced , Muscular Atrophy/physiopathology , Physical Conditioning, Animal/physiology , Animals , Exercise Test/methods , Gene Expression/drug effects , Gene Expression/physiology , Male , Mice , Mice, Inbred C57BL , Muscle Proteins/metabolism , Muscle, Skeletal/drug effects , Muscle, Skeletal/metabolism , Muscular Atrophy/metabolism , Myostatin/metabolism , Phosphorylation/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Ribosomal Protein S6 Kinases, 70-kDa/metabolism , TOR Serine-Threonine Kinases/metabolism , Up-Regulation/drug effects , Up-Regulation/physiology
18.
Inflamm Res ; 66(8): 663-678, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28405735

ABSTRACT

OBJECTIVES: Contact dermatitis model involving repeated application of hapten is used as a tool to assess dermatitis, as characterized by thickening. Involvement of cell proliferation, elicited by repeated hapten-stimulation, in this swelling has been unclear. Curcumin is reported to reduce inflammation. We examined involvement of cell proliferation and the role of extracellular regulated kinase (ERK) in 2,4,6-trinitrochlorobenzene (TNCB) challenge-induced ear swelling. We also examined the effects of curcumin in this model. METHODS: Mice were sensitized with TNCB to the abdominal skin. Then, they were challenged with TNCB to the ear three times. The ERK activation inhibitor U0126 or curcumin was applied 30 min before each TNCB challenge. RESULTS: TNCB challenge-induced increased epidermal cell number and dermal thickening. Gene expressions of epithelial mitogen (EPGN), amphiregulin (AREG) and heparin-binding-epidermal growth factor (HB-EGF) were increased in the ears after the last TNCB challenge. Ki-67 immunoreactivity was increased in the dermis in TNCB-challenged ears. TNCB-induced swelling was inhibited by U0126 and curcumin. Curcumin also attenuated TNCB-induced ERK phosphorylation and expression of EPGN and AREG genes. CONCLUSION: Ear swelling induced by TNCB challenge might be mediated, in part, by the EPGN- and AREG-ERK proliferation pathway and was inhibited by curcumin.


Subject(s)
Amphiregulin/metabolism , Curcumin/pharmacology , Dermatitis, Allergic Contact/metabolism , Epigen/metabolism , Animals , Cytokines/genetics , Dermatitis, Allergic Contact/pathology , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Haptens , Intercellular Signaling Peptides and Proteins/genetics , Ki-67 Antigen/genetics , Ki-67 Antigen/metabolism , Mice, Inbred BALB C , Phosphorylation/drug effects , Picryl Chloride , Skin/drug effects , Skin/metabolism , Skin/pathology , Up-Regulation/drug effects
20.
Biol Pharm Bull ; 39(1): 33-41, 2016.
Article in English | MEDLINE | ID: mdl-26567724

ABSTRACT

Recently the global expression of taste 2 receptors (TAS2Rs) on smooth muscle cells in human airways was demonstrated. Here, the effects of agonists of taste receptor, type 2, denatonium and 6-n-propyl-2-thiouracil, on smooth-muscle contraction were examined in the rat and mouse. Contractions induced by carbachol (CCh), high K(+), and sodium fluoride, but not calyculin-A, were inhibited significantly in the presence of a TAS2R agonist in the bronchial smooth muscle of mice. The contraction induced by CCh was inhibited by TAS2R agonists in ileal smooth muscle. Phenylephrine-induced contraction was also inhibited by TAS2R agonists in aortic smooth muscle. Gastrointestinal motility and blood pressure were attenuated by administration of TAS2R agonists in vivo. These findings suggest that TAS2R may be receptor for endogenous biologically active substances as well as for bitter tastes on the tongue. TAS2R signaling could be employed in the development of anti-asthmatic, anti-spasmodic, and anti-hypertensive drugs.


Subject(s)
Muscle Contraction/drug effects , Muscle, Smooth/drug effects , Propylthiouracil/pharmacology , Quaternary Ammonium Compounds/pharmacology , Receptors, G-Protein-Coupled/agonists , Animals , Carbachol/pharmacology , Humans , Male , Mice , Mice, Inbred C57BL , Potassium/pharmacology , Rats , Sodium Fluoride/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL