Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 100
Filter
1.
Biomed Pharmacother ; 143: 112188, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34563947

ABSTRACT

An extract from Artemisia dracunculus L. (termed PMI-5011) improves glucose homeostasis by enhancing insulin action and reducing ectopic lipid accumulation, while increasing fat oxidation in skeletal muscle tissue in obese insulin resistant male mice. A chalcone, DMC-2, in PMI-5011 is the major bioactive that enhances insulin signaling and activation of AKT. However, the mechanism by which PMI-5011 improves lipid metabolism is unknown. AMPK is the cellular energy and metabolic sensor and a key regulator of lipid metabolism in muscle. This study examined PMI-5011 activation of AMPK signaling using murine C2C12 muscle cell culture and skeletal muscle tissue. Findings show that PMI-5011 increases Thr172-phosphorylation of AMPK in muscle cells and skeletal muscle tissue, while hepatic AMPK activation by PMI-5011 was not observed. Increased AMPK activity by PMI-5011 affects downstream signaling of AMPK, resulting in inhibition of ACC and increased SIRT1 protein levels. Selective deletion of DMC-2 from PMI-5011 demonstrates that compounds other than DMC-2 in a "DMC-2 knock out extract" (KOE) are responsible for AMPK activation and its downstream effects. Compared to 5-aminoimidazole-4-carboxamide ribonucleotide (AICAR) and metformin, the phytochemical mixture characterizing the KOE appears to more efficiently activate AMPK in muscle cells. KOE-mediated AMPK activation was LKB-1 independent, suggesting KOE does not activate AMPK via LKB-1 stimulation. Through AMPK activation, compounds in PMI-5011 may regulate lipid metabolism in skeletal muscle. Thus, the AMPK-activating potential of the KOE adds therapeutic value to PMI-5011 and its constituents in treating insulin resistance or type 2 diabetes.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Artemisia , Enzyme Activators/pharmacology , Hypoglycemic Agents/pharmacology , Insulin Resistance , Muscle, Skeletal/drug effects , Phytochemicals/pharmacology , Plant Extracts/pharmacology , Aminoimidazole Carboxamide/analogs & derivatives , Aminoimidazole Carboxamide/pharmacology , Animals , Artemisia/chemistry , Cell Line , Diet, High-Fat , Disease Models, Animal , Enzyme Activation , Enzyme Activators/isolation & purification , Hypoglycemic Agents/isolation & purification , Male , Metformin/pharmacology , Mice, Inbred C57BL , Muscle, Skeletal/enzymology , Myoblasts, Skeletal/drug effects , Myoblasts, Skeletal/enzymology , Phosphorylation , Phytochemicals/isolation & purification , Plant Extracts/isolation & purification , Ribonucleotides/pharmacology , Signal Transduction/drug effects
2.
Int J Biol Macromol ; 187: 603-613, 2021 Sep 30.
Article in English | MEDLINE | ID: mdl-34314795

ABSTRACT

Although crotoxin B (CB) is a well-established catalytically active secretory phospholipase A2 group IIA (sPLA2-IIA) myotoxin, we investigated its potential stimulatory effect on myogenesis with the involvement of prostaglandins (PGs) produced by cyclooxygenase (COX)-1 and -2 pathways. Myoblast C2C12 were cultured in proliferation or commitment protocols and incubated with CB followed by lumiracoxib (selective COX-2 inhibitor) or valeryl salicylate (selective COX-1 inhibitor) and subjected to analysis of PG release, cell proliferation and activation of myogenic regulatory factors (MRFs). Our data showed that CB in non-cytotoxic concentrations induces an increase of COX-2 protein expression and stimulates the activity of both COX isoforms to produce PGE2, PGD2 and 15d-PGJ2. CB induced an increase in the proliferation of C2C12 myoblast cells dependent on PGs from both COX-1 and COX-2 pathways. In addition, CB stimulated the activity of Pax7, MyoD, Myf5 and myogenin in proliferated cells. Otherwise, CB increased myogenin activity but not MyoD in committed cells. Our findings evidence the role of COX-1- and COX-2-derived PGs in modulating CB-induced activation of MRFs. This study contributes to the knowledge that CB promote early myogenic events via regulatory mechanisms on PG-dependent COX pathways, showing new concepts about the effect of sPLA2-IIA in skeletal muscle repair.


Subject(s)
Cell Differentiation/drug effects , Cell Proliferation/drug effects , Crotoxin/pharmacology , Cyclooxygenase 1/metabolism , Cyclooxygenase 2/metabolism , Group II Phospholipases A2/pharmacology , Membrane Proteins/metabolism , Muscle Development/drug effects , Myoblasts, Skeletal/drug effects , Neurotoxins/pharmacology , Prostaglandins/metabolism , Animals , Cell Line , Mice , MyoD Protein/metabolism , Myoblasts, Skeletal/enzymology , Myogenic Regulatory Factor 5/metabolism , Myogenin/metabolism , PAX7 Transcription Factor/metabolism , Signal Transduction
3.
Molecules ; 26(6)2021 Mar 12.
Article in English | MEDLINE | ID: mdl-33809377

ABSTRACT

Muscle fatigue is induced by an acute or chronic physical performance inability after excessive physical activity often associated with lactate accumulation, the end-product of glycolysis. In this study, the water-extracted roots of Sanguisorba officinalis L., a herbal medicine traditionally used for inflammation and diarrhea, reduced the activities of lactate dehydrogenase A (LDHA) in in vitro enzyme assay myoblast C2C12 cells and murine muscle tissue. Physical performance measured by a treadmill test was improved in the S. officinalis-administrated group. The analysis of mouse serum and tissues showed significant changes in lactate levels. Among the proteins related to energy metabolism-related physical performance, phosphorylated-AMP-activated protein kinase alpha (AMPKα) and peroxisome proliferator-activated receptor-coactivator-1 alpha (PGC-1α) levels were enhanced, whereas the amount of LDHA was suppressed. Therefore, S. officinalis might be a candidate for improving physical performance via inhibiting LDHA and glycolysis.


Subject(s)
Lactate Dehydrogenase 5/antagonists & inhibitors , Physical Functional Performance , Plant Extracts/administration & dosage , Plants, Medicinal/chemistry , Sanguisorba/chemistry , AMP-Activated Protein Kinases/metabolism , Administration, Oral , Animals , Cell Line , Exercise Test , Glycolysis/drug effects , Lactic Acid/metabolism , Male , Medicine, Korean Traditional , Mice , Mice, Inbred C57BL , Myoblasts, Skeletal/drug effects , Myoblasts, Skeletal/enzymology , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Physical Endurance/drug effects , Phytochemicals/administration & dosage , Phytochemicals/chemistry , Phytotherapy , Plant Extracts/chemistry
4.
Thyroid ; 31(1): 115-127, 2021 01.
Article in English | MEDLINE | ID: mdl-32787533

ABSTRACT

Background: The type 2 deiodinase (DIO2) converts thyroxine to 3,3',5-triiodothyronine (T3), modulating intracellular T3. An increase in DIO2 within muscle stem cells during skeletal muscle regeneration leads to T3-dependent potentiation of differentiation. The muscle stem cell niche comprises numerous cell types, which coordinate the regeneration process. For example, muscle stem cells provide secretory signals stimulating endothelial cell-mediated vascular repair, and, in turn, endothelial cells promote muscle stem differentiation. We hypothesized that Dio2 loss in muscle stem cells directly impairs muscle stem cell-endothelial cell communication, leading to downstream disruption of endothelial cell function. Methods: We assessed the production of proangiogenic factors in differentiated C2C12 cells and in a C2C12 cell line without Dio2 (D2KO C2C12) by real-time quantitative-polymerase chain reaction and enzyme-linked immunosorbent assay. Conditioned medium (CM) was collected daily in parallel to evaluate its effects on human umbilical vein endothelial cell (HUVEC) proliferation, migration and chemotaxis, and vascular network formation. The effects of T3-treatment on vascular endothelial growth factor (Vegfa) mRNA expression in C2C12 cells and mouse muscle were assessed. Chromatin immunoprecipitation (ChIP) identified thyroid hormone receptor (TR) binding to the Vegfa gene. Using mice with a targeted disruption of Dio2 (D2KO mice), we determined endothelial cell number by immunohistochemistry/flow cytometry and evaluated related gene expression in both uninjured and injured skeletal muscle. Results: In differentiated D2KO C2C12 cells, Vegfa expression was 46% of wildtype (WT) C2C12 cells, while secreted VEGF was 45%. D2KO C2C12 CM exhibited significantly less proangiogenic effects on HUVECs. In vitro and in vivo T3 treatment of C2C12 cells and WT mice, and ChIP using antibodies against TRα, indicated that Vegfa is a direct genomic T3 target. In uninjured D2KO soleus muscle, Vegfa expression was decreased by 28% compared with WT mice, while endothelial cell numbers were decreased by 48%. Seven days after skeletal muscle injury, D2KO mice had 36% fewer endothelial cells, coinciding with an 83% decrease in Vegfa expression in fluorescence-activated cell sorting purified muscle stem cells. Conclusion:Dio2 loss in the muscle stem cell impairs muscle stem cell-endothelial cell crosstalk via changes in the T3-responsive gene Vegfa, leading to downstream impairment of endothelial cell function both in vitro and in vivo.


Subject(s)
Human Umbilical Vein Endothelial Cells/metabolism , Iodide Peroxidase/metabolism , Muscle Development , Muscle, Skeletal/enzymology , Myoblasts, Skeletal/enzymology , Neovascularization, Physiologic , Paracrine Communication , Regeneration , Vascular Endothelial Growth Factor A/metabolism , Animals , Cell Line , Cell Movement , Cell Proliferation , Humans , Iodide Peroxidase/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Muscle, Skeletal/injuries , Muscle, Skeletal/pathology , Myoblasts, Skeletal/pathology , Signal Transduction , Up-Regulation , Vascular Endothelial Growth Factor A/genetics , Iodothyronine Deiodinase Type II
5.
FASEB J ; 35(1): e21154, 2021 01.
Article in English | MEDLINE | ID: mdl-33140469

ABSTRACT

Myogenesis includes sequential stages of progenitor cell proliferation, myogenic commitment and differentiation, myocyte fusion, and myotube maturation. Different stages of myogenesis are orchestrated and regulated by myogenic regulatory factors and various downstream cellular signaling. Here we identify phosphatase orphan 1 (Phospho1) as a new player in myogenesis. During activation, proliferation, and differentiation of quiescent satellite cells, the expression of Phospho1 gradually increases. Overexpression of Phospho1 inhibits myoblast proliferation but promotes their differentiation and fusion. Conversely, knockdown of Phospho1 accelerates myoblast proliferation but impairs myotube formation. Moreover, knockdown of Phospho1 decreases the OXPHO protein levels and mitochondria density, whereas overexpression of Phospho1 upregulates OXPHO protein levels and promotes mitochondrial oxygen consumption. Finally, we show that Phospho1 expression is controlled by myogenin, which binds to the promoter of Phospho1 to regulate its transcription. These results indicate a key role of Phospho1 in regulating myogenic differentiation and mitochondrial function.


Subject(s)
Cell Differentiation , Cell Proliferation , Gene Expression Regulation, Enzymologic , Muscle Development , Myoblasts, Skeletal/enzymology , Phosphoric Monoester Hydrolases/biosynthesis , Animals , Mice , Mitochondria, Muscle/genetics , Mitochondria, Muscle/metabolism , Myogenin/genetics , Myogenin/metabolism , Phosphoric Monoester Hydrolases/genetics
6.
FASEB J ; 34(9): 12946-12962, 2020 09.
Article in English | MEDLINE | ID: mdl-32772437

ABSTRACT

Although we have shown that catecholamines suppress the activity of the Ubiquitin-Proteasome System (UPS) and atrophy-related genes expression through a cAMP-dependent manner in skeletal muscle from rodents, the underlying mechanisms remain unclear. Here, we report that a single injection of norepinephrine (NE; 1 mg kg-1 ; s.c) attenuated the fasting-induced up-regulation of FoxO-target genes in tibialis anterior (TA) muscles by the stimulation of PKA/CREB and Akt/FoxO1 signaling pathways. In addition, muscle-specific activation of PKA by the overexpression of PKA catalytic subunit (PKAcat) suppressed FoxO reporter activity induced by (1) a wild-type; (2) a non-phosphorylatable; (3) a non-phosphorylatable and non-acetylatable forms of FoxO1 and FoxO3; (4) downregulation of FoxO protein content, and probably by (5) PGC-1α up-regulation. Consistently, the overexpression of the PKAcat inhibitor (PKI) up-regulated FoxO activity and the content of Atrogin-1 and MuRF1, as well as induced muscle fiber atrophy, the latter effect being prevented by the overexpression of a dominant negative (d. n.) form of FoxO (d.n.FoxO). The sustained overexpression of PKAcat induced fiber-type transition toward a smaller, slower, and more oxidative phenotype and improved muscle resistance to fatigue. Taken together, our data provide the first evidence that endogenous PKA activity is required to restrain the basal activity of FoxO and physiologically important to maintain skeletal muscle mass.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/metabolism , Forkhead Box Protein O1/metabolism , Muscle, Skeletal/enzymology , Muscular Atrophy/metabolism , Animals , Cell Line , Forkhead Box Protein O3/metabolism , Male , Mice , Mice, Inbred C57BL , Muscle, Skeletal/pathology , Myoblasts, Skeletal/enzymology , Signal Transduction
7.
Biomed Pharmacother ; 128: 110238, 2020 Aug.
Article in English | MEDLINE | ID: mdl-32450522

ABSTRACT

Muscle atrophy is the loss of skeletal muscle mass during several pathological conditions such as long-term fasting, aging, cancer, diabetes, sepsis and immune disorders. Glucocorticoids are known to trigger skeletal muscle atrophy. Dexamethasone (DEX), a synthetic glucocorticoid, induces skeletal muscle atrophy by suppression of protein synthesis and promotion of protein degradation. The double-stranded RNA (dsRNA)-activated protein kinase R (PKR) plays a significant role in mediating lipopolysaccharide-induced inflammation. However, pathological roles of PKR in muscle atrophy are not fully understood. The current study aimed to investigate the effect of imoxin, a PKR inhibitor, on DEX-induced muscle atrophy in C2C12 myotubes. Myotubes were incubated with imoxin at different concentrations with or without 5 µM DEX for 24 h. In the current study, imoxin treatment significantly reduced protein levels of MuRF1 and MAFbx induced by DEX by 88 ± 2% and MAFbx by 99 ± 0%, respectively. Moreover, 5 µM imoxin treatment reduced protein ubiquitination by 42 ± 4% and protein content of nuclear FoxO3α (77 ± 4%) in presence of DEX. Furthermore, 5 µM imoxin treatment stimulated Akt phosphorylation (195 ± 5%), mTOR phosphorylation (171 ± 21 %) and p70S6K1 phosphorylation (314 ± 31 %) under DEX-treated condition even though DEX treatment did not suppressed Akt/mTOR/p70S6K1 axis. These findings suggest that imoxin may protect against DEX-induced skeletal muscle atrophy by alleviating muscle specific E3 ubiquitin ligases and imoxin alone may promote protein synthesis via Akt/mTOR/S6K1 axis in muscle cells.


Subject(s)
Anabolic Agents/pharmacology , Dexamethasone/toxicity , Imidazoles/pharmacology , Indoles/pharmacology , Muscular Atrophy/prevention & control , Myoblasts, Skeletal/drug effects , Protein Kinase Inhibitors/pharmacology , Ubiquitin-Protein Ligases/metabolism , eIF-2 Kinase/antagonists & inhibitors , Animals , Cell Line , Forkhead Box Protein O3/metabolism , Mice , Muscle Proteins/metabolism , Muscular Atrophy/chemically induced , Muscular Atrophy/enzymology , Muscular Atrophy/pathology , Myoblasts, Skeletal/enzymology , Myoblasts, Skeletal/pathology , Phosphorylation , Proteasome Endopeptidase Complex/drug effects , Proteasome Endopeptidase Complex/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Ribosomal Protein S6 Kinases, 70-kDa/metabolism , SKP Cullin F-Box Protein Ligases/metabolism , Signal Transduction , TOR Serine-Threonine Kinases/metabolism , Tripartite Motif Proteins/metabolism , eIF-2 Kinase/metabolism
8.
Eur J Pharmacol ; 881: 173215, 2020 Aug 15.
Article in English | MEDLINE | ID: mdl-32473166

ABSTRACT

Insulin resistance (IR) is a constituent part of Type 2 Diabetes Mellitus (T2DM). Conditioned medium from Adipose derived Mesenchymal Stem Cells (ADMSCs-CM) has been shown to reverse IR. However, its effect on cellular stress is not well established. The objective of this study was to explore the effect of ADMSCs-CM on reactive oxygen species, mitochondrial membrane potential (ΔΨm), endoplasmic reticulum (ER) stress and expression of oxidative and inflammatory stress induced serine kinases (SISK) which are pathophysiologically linked to IR. In insulin resistant, 3T3-L1 adipocytes and C2C12 myoblast cell culture models, glucose uptake was assayed by 2-NBDG uptake. Immunomodulatory cytokines, intracellular reactive oxygen species generation, ΔΨm and protein expression of JNK1, IKKß and phospho-IRS1 (307) were analyzed using FACS. mRNA expression of ER stress markers (CHOP1 and IRE1) and SISK (JNK1, IKKß, ERK1 and S6K1) were analyzed using RT-PCR. ADMSCs-CM effectively improve glucose uptake as evidenced by 2-NBDG uptake assay. FACS analysis showed that ADMSCs-CM possessed significantly higher levels of IL-6 and IL-10. ADMSCs-CM decreased intracellular generation of reactive oxygen species where it restored ΔΨm in C2C12 cells. ADMSCs-CM mediated reduction in ER stress was confirmed by down-regulation in CHOP1 and IRE1 mRNA expression. ADMSCs-CM treatment showed significant down-regulation of SISK mRNA expression including IKKß, JNK, ERK and S6K1. Our results unequivocally demonstrate for the first time the mechanism of action of ADMSCs-CM in amelioration IR by reducing oxidative and inflammatory cellular stress. This study identifies SISK as potential therapeutic targets for T2DM therapy.


Subject(s)
Culture Media, Conditioned/metabolism , Diabetes Mellitus, Type 2/enzymology , Insulin Resistance , Mesenchymal Stem Cells/metabolism , Myoblasts, Skeletal/enzymology , Paracrine Communication , Protein Kinases/metabolism , Stress, Physiological , 3T3-L1 Cells , Adipose Tissue/cytology , Animals , Cytokines/metabolism , Endoplasmic Reticulum Stress , Gene Expression Regulation, Enzymologic , Inflammation Mediators/metabolism , Insulin Receptor Substrate Proteins/metabolism , Membrane Potential, Mitochondrial , Mice , Oxidative Stress , Phosphorylation , Protein Kinases/genetics , Reactive Oxygen Species/metabolism , Signal Transduction
9.
Drug Chem Toxicol ; 43(6): 637-644, 2020 Nov.
Article in English | MEDLINE | ID: mdl-30426790

ABSTRACT

This study was designed to investigate possible interference of Xenobiotics with SUMOylation in eukaryotic cells. To begin with, we docked 71 chemical structures from PubChem with human SUMO1 and UBC9 protein structures using Auto Dock 4.2 and Hex 6.3 and selected five compounds for binding studies in Surface Plasmon Resonance (SPR) with human SUMO1. In SPR studies, only endosulfan showed binding to SUMO1 (Kd1.313 × 10-4 M). Further, we treated HePG2 and differentiated 3T3-L1 cells with endosulfan/bisphenol A/perfluorooctanoic acid (PFOA) to test induction of oxidative stress and SUMO isoform/UBC9 expression. Treatment with these compounds resulted in higher levels of nitric oxide (NO), NOS2A mRNA, and reactive oxygen species (ROS) associated with decreased NADPH levels. Additionally, treatment with these chemicals resulted in elevated mRNA levels of IL-6 and IL-1ß in 3T3-L1 cells. In HePG2 cells, endosulfan treatment resulted in elevated mRNA levels of SUMO1, 3 and UBC9, whereas, treatment with bisphenol A resulted in increased mRNA of SUMO2, 3 and UBC9. Treatment with PFOA resulted in elevated mRNA levels of SUMO2. Apart from influencing the gene expression, endosulfan caused decrease in SUMO1-Sumoylation of few proteins. We propose that one reason for the severe health consequences of exposure to endosulfan/bisphenol could be due to induction of oxidative stress and modulation in SUMO and UBC9 gene expression.


Subject(s)
Adipocytes/drug effects , Benzhydryl Compounds/toxicity , Endosulfan/toxicity , Hepatocytes/drug effects , Myoblasts, Skeletal/drug effects , Phenols/toxicity , SUMO-1 Protein/metabolism , Small Ubiquitin-Related Modifier Proteins/metabolism , Ubiquitin-Conjugating Enzymes/metabolism , Ubiquitins/metabolism , 3T3-L1 Cells , Adipocytes/enzymology , Adipocytes/pathology , Animals , Benzhydryl Compounds/metabolism , Endosulfan/metabolism , Hep G2 Cells , Hepatocytes/enzymology , Hepatocytes/pathology , Humans , Mice , Molecular Docking Simulation , Myoblasts, Skeletal/enzymology , Myoblasts, Skeletal/pathology , Oxidative Stress/drug effects , Phenols/metabolism , Protein Binding , Reactive Oxygen Species/metabolism , SUMO-1 Protein/genetics , Small Ubiquitin-Related Modifier Proteins/genetics , Sumoylation , Ubiquitin-Conjugating Enzymes/genetics , Ubiquitins/genetics
10.
FASEB J ; 33(10): 10648-10667, 2019 10.
Article in English | MEDLINE | ID: mdl-31268746

ABSTRACT

Casein kinase 2 (CK2) is a tetrameric protein kinase composed of 2 catalytic (α and α') and 2 regulatory ß subunits. Our study provides the first molecular and cellular characterization of the different CK2 subunits, highlighting their individual roles in skeletal muscle specification and differentiation. Analysis of C2C12 cell knockout for each CK2 subunit reveals that: 1) CK2ß is mandatory for the expression of the muscle master regulator myogenic differentiation 1 in proliferating myoblasts, thus controlling both myogenic commitment and subsequent muscle-specific gene expression and myotube formation; 2) CK2α is involved in the activation of the muscle-specific gene program; and 3) CK2α' activity regulates myoblast fusion by mediating plasma membrane translocation of fusogenic proteins essential for membrane coalescence, like myomixer. Accordingly, CK2α' overexpression in C2C12 cells and in mouse regenerating muscle is sufficient to increase myofiber size and myonuclei content via enhanced satellite cell fusion. Consistent with these results, pharmacological inhibition of CK2 activity substantially blocks the expression of myogenic markers and muscle cell fusion both in vitro in C2C12 and primary myoblasts and in vivo in mouse regenerating muscle and zebrafish development. Overall, our work describes the specific and coordinated functions of CK2 subunits in orchestrating muscle differentiation and fusogenic activity, highlighting CK2 relevance in the physiopathology of skeletal muscle tissue.-Salizzato, V., Zanin, S., Borgo, C., Lidron, E., Salvi, M., Rizzuto, R., Pallafacchina, G., Donella-Deana, A. Protein kinase CK2 subunits exert specific and coordinated functions in skeletal muscle differentiation and fusogenic activity.


Subject(s)
Casein Kinase II/physiology , Muscle, Skeletal/cytology , Muscle, Skeletal/enzymology , Animals , Casein Kinase II/antagonists & inhibitors , Casein Kinase II/genetics , Cell Differentiation/genetics , Cell Differentiation/physiology , Cell Fusion , Cell Line , Gene Knockout Techniques , Male , Mice , Mice, Inbred C57BL , Models, Biological , Muscle Development/genetics , Muscle Development/physiology , MyoD Protein/genetics , MyoD Protein/metabolism , Myoblasts, Skeletal/cytology , Myoblasts, Skeletal/enzymology , Protein Subunits , Satellite Cells, Skeletal Muscle/cytology , Satellite Cells, Skeletal Muscle/enzymology , Zebrafish , Zebrafish Proteins/antagonists & inhibitors , Zebrafish Proteins/genetics , Zebrafish Proteins/physiology
11.
J Cell Biochem ; 120(11): 18854-18861, 2019 11.
Article in English | MEDLINE | ID: mdl-31257635

ABSTRACT

Cdk9 is a serine-threonine protein kinase that has been recognized as a regulator of cardiac differentiation. Recently, we have reported that transient induction of Cdk9 using noncoding RNA targeting Cdk9 sequences results in efficient cardiac differentiation. Concerning Cdk9 regulatory roles, here, we proposed whether constant overexpression of Cdk9 might influence the differentiation of myoblast C2C12 cells into myotubes. We overexpressed Cdk9 in mouse myoblast C2C12 cells to investigate its regulatory roles on myogenic differentiation. Upon Cdk9 overexpression, the expression level of myogenic regulatory factors was determined. Moreover, the expression profile of three important myomiRs consist of miR 1, 133 and 206 was examined during the differentiation process. Although Cdk9 expression is necessary for inducing differentiation in the early stage of myogenesis, continuous Cdk9 expression inhibits differentiation by modulating myomiRs and myogenic gene expression. Our results indicate that the transient induction of Cdk9 in the early stage of differentiation is critical for myogenesis.


Subject(s)
Cell Differentiation , Cyclin-Dependent Kinase 9/biosynthesis , Muscle Development , Muscle Fibers, Skeletal/enzymology , Myoblasts, Skeletal/enzymology , Animals , Cell Line , Cyclin-Dependent Kinase 9/genetics , Enzyme Induction , Mice , MicroRNAs/genetics , MicroRNAs/metabolism , Muscle Fibers, Skeletal/cytology , Myoblasts, Skeletal/cytology
12.
Am J Physiol Cell Physiol ; 314(6): C721-C731, 2018 06 01.
Article in English | MEDLINE | ID: mdl-29513566

ABSTRACT

Unloading-mediated muscle atrophy is associated with increased reactive oxygen species (ROS) production. We previously demonstrated that elevated ubiquitin ligase casitas B-lineage lymphoma-b (Cbl-b) resulted in the loss of muscle volume (Nakao R, Hirasaka K, Goto J, Ishidoh K, Yamada C, Ohno A, Okumura Y, Nonaka I, Yasutomo K, Baldwin KM, Kominami E, Higashibata A, Nagano K, Tanaka K, Yasui N, Mills EM, Takeda S, Nikawa T. Mol Cell Biol 29: 4798-4811, 2009). However, the pathological role of ROS production associated with unloading-mediated muscle atrophy still remains unknown. Here, we showed that the ROS-mediated signal transduction caused by microgravity or its simulation contributes to Cbl-b expression. In L6 myotubes, the assessment of redox status revealed that oxidized glutathione was increased under microgravity conditions, and simulated microgravity caused a burst of ROS, implicating ROS as a critical upstream mediator linking to downstream atrophic signaling. ROS generation activated the ERK1/2 early-growth response protein (Egr)1/2-Cbl-b signaling pathway, an established contributing pathway to muscle volume loss. Interestingly, antioxidant treatments such as N-acetylcysteine and TEMPOL, but not catalase, blocked the clinorotation-mediated activation of ERK1/2. The increased ROS induced transcriptional activity of Egr1 and/or Egr2 to stimulate Cbl-b expression through the ERK1/2 pathway in L6 myoblasts, since treatment with Egr1/2 siRNA and an ERK1/2 inhibitor significantly suppressed clinorotation-induced Cbl-b and Egr expression, respectively. Promoter and gel mobility shift assays revealed that Cbl-b was upregulated via an Egr consensus oxidative responsive element at -110 to -60 bp of the Cbl-b promoter. Together, this indicates that under microgravity conditions, elevated ROS may be a crucial mechanotransducer in skeletal muscle cells, regulating muscle mass through Cbl-b expression activated by the ERK-Egr signaling pathway.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Muscular Atrophy/enzymology , Myoblasts, Skeletal/enzymology , Oxidative Stress , Proto-Oncogene Proteins c-cbl/metabolism , Reactive Oxygen Species/metabolism , Weightlessness , Adaptor Proteins, Signal Transducing/genetics , Animals , Antioxidants/pharmacology , COS Cells , Chlorocebus aethiops , Early Growth Response Transcription Factors/genetics , Early Growth Response Transcription Factors/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Glutathione/metabolism , Mechanotransduction, Cellular , Muscular Atrophy/genetics , Muscular Atrophy/pathology , Muscular Atrophy/prevention & control , Myoblasts, Skeletal/drug effects , Myoblasts, Skeletal/pathology , Oxidation-Reduction , Oxidative Stress/drug effects , Phosphorylation , Promoter Regions, Genetic , Proto-Oncogene Proteins c-cbl/genetics , Rats , Space Flight , Time Factors , Up-Regulation , Weightlessness Simulation
13.
Physiol Rep ; 6(6): e13652, 2018 03.
Article in English | MEDLINE | ID: mdl-29595878

ABSTRACT

Skeletal muscle physiology and metabolism are regulated by complex networks of intracellular signaling pathways. Among many of these pathways, the protein kinase AKT plays a prominent role. While three AKT isoforms have been identified (AKT1, AKT2, and AKT3), surprisingly little is known regarding isoform-specific expression of AKT in human skeletal muscle. To address this, we examined the expressions of each AKT isoform in muscle biopsy samples collected from the vastus lateralis of healthy male adults at rest. In muscle, AKT2 was the most highly expressed AKT transcript, exhibiting a 15.4-fold increase over AKT1 and AKT3 transcripts. Next, the abundance of AKT protein isoforms was determined using antibody immunoprecipitation followed by Liquid Chromatography-Parallel Reaction Monitoring/Mass Spectrometry. Immunoprecipitation was performed using either mouse or rabbit pan AKT antibodies that were immunoreactive with all three AKT isoforms. We found that AKT2 was the most abundant AKT isoform in human skeletal muscle (4.2-fold greater than AKT1 using the rabbit antibody and 1.6-fold greater than AKT1 using the mouse antibody). AKT3 was virtually undetectable. Next, cultured primary human myoblasts were virally-transduced with cDNAs encoding either wild-type (WT) or kinase-inactive AKT1 (AKT1-K179M) or AKT2 (AKT2-K181M) and allowed to terminally differentiate. Myotubes expressing WT-AKT1 or WT-AKT2 showed enhanced fusion compared to control myotubes, while myotubes expressing AKT1-K179M showed a 14% reduction in fusion. Myotubes expressing AKT2-K181M displayed 63% decreased fusion compared to control. Together, these data identify AKT2 as the most highly-expressed AKT isoform in human skeletal muscle and as the principal AKT isoform regulating human myoblast differentiation.


Subject(s)
Muscle, Skeletal/enzymology , Proto-Oncogene Proteins c-akt/biosynthesis , Adult , Cell Differentiation/physiology , Humans , Isoenzymes , Male , Myoblasts, Skeletal/cytology , Myoblasts, Skeletal/enzymology
14.
Skelet Muscle ; 8(1): 5, 2018 02 20.
Article in English | MEDLINE | ID: mdl-29463296

ABSTRACT

BACKGROUND: Tyrosine kinase inhibitors (TKIs) are effective therapies with demonstrated antineoplastic activity. Nilotinib is a second-generation FDA-approved TKI designed to overcome Imatinib resistance and intolerance in patients with chronic myelogenous leukemia (CML). Interestingly, TKIs have also been shown to be an efficient treatment for several non-malignant disorders such fibrotic diseases, including those affecting skeletal muscles. METHODS: We investigated the role of Nilotinib on skeletal myogenesis using the well-established C2C12 myoblast cell line. We evaluated the impact of Nilotinib during the time course of skeletal myogenesis. We compared the effect of Nilotinib with the well-known p38 MAPK inhibitor SB203580. MEK1/2 UO126 and PI3K/AKT LY294002 inhibitors were used to identify the signaling pathways involved in Nilotinib-related effects on myoblast. Adult primary myoblasts were also used to corroborate the inhibition of myoblasts fusion and myotube-nuclei positioning by Nilotinib. RESULTS: We found that Nilotinib inhibited myogenic differentiation, reducing the number of myogenin-positive myoblasts and decreasing myogenin and MyoD expression. Furthermore, Nilotinib-mediated anti-myogenic effects impair myotube formation, myosin heavy chain expression, and compromise myotube-nuclei positioning. In addition, we found that p38 MAPK is a new off-target protein of Nilotinib, which causes inhibition of p38 phosphorylation in a similar manner as the well-characterized p38 inhibitor SB203580. Nilotinib induces the activation of ERK1/2 and AKT on myoblasts but not in myotubes. We also found that Nilotinib stimulates myoblast proliferation, a process dependent on ERK1/2 and AKT activation. CONCLUSIONS: Our findings suggest that Nilotinib may have important negative effects on muscle homeostasis, inhibiting myogenic differentiation but stimulating myoblasts proliferation. Additionally, we found that Nilotinib stimulates the activation of ERK1/2 and AKT. On the other hand, we suggest that p38 MAPK is a new off-target of Nilotinib. Thus, there is a necessity for future studies to investigate the long-term effects of TKIs on skeletal muscle homeostasis, along with potential detrimental effects in cell differentiation and proliferation in patients receiving TKI therapies.


Subject(s)
Muscle Development/drug effects , Myoblasts, Skeletal/drug effects , Protein Kinase Inhibitors/pharmacology , Pyrimidines/pharmacology , Animals , Apoptosis/drug effects , Apoptosis/physiology , Cell Differentiation/drug effects , Cell Differentiation/physiology , Cell Proliferation/drug effects , Cell Proliferation/physiology , Cells, Cultured , Gene Expression Regulation/drug effects , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/physiology , Mice, Inbred C57BL , Muscle Development/physiology , Muscle Fibers, Skeletal/cytology , Muscle Fibers, Skeletal/drug effects , Muscle Fibers, Skeletal/metabolism , Myoblasts, Skeletal/cytology , Myoblasts, Skeletal/enzymology , Myogenin/biosynthesis , Myogenin/genetics , Phosphorylation/drug effects , Phosphorylation/physiology , Protein-Tyrosine Kinases/antagonists & inhibitors , Proteostasis/drug effects , Proteostasis/physiology , Proto-Oncogene Proteins c-akt/metabolism , RNA, Messenger/genetics , p38 Mitogen-Activated Protein Kinases/metabolism
15.
Stem Cell Res Ther ; 9(1): 33, 2018 02 09.
Article in English | MEDLINE | ID: mdl-29426367

ABSTRACT

BACKGROUND: Skeletal muscle satellite cell-derived myoblasts are mainly responsible for postnatal muscle growth and injury-induced regeneration. Many intracellular signaling pathways are essential for myogenic differentiation, while a number of kinases are involved in this modulation process. Type I phosphatidylinositol 4-phosphate 5-kinase (PIP5KI) was identified as one of the key kinases involved in myogenic differentiation, but the underlying molecular mechanism is still unclear. METHODS: PIP5K1α was quantified by quantitative reverse transcriptase PCR and western blot assay. Expression levels of myogenin and myosin heavy chain, which showed significant downregulation in PIP5K1α siRNA-mediated knockdown cells in western blot analysis, were confirmed by immunostaining. Phosphatidylinositol 4,5-bisphosphate in PIP5K1α siRNA-mediated knockdown cells was also measured by the PI(4,5)P2 Mass ELISA Kit. C2C12 cells were overexpressed with different forms of AKT, followed by western blot analysis on myogenin and myosin heavy chain, which reveals their function in myogenic differentiation. FLIPR assays are used to test the release of calcium in PIP5K1α siRNA-mediated knockdown cells after histamine or bradykinin treatment. Statistical significances between groups were determined by two-tailed Student's t test. RESULTS: Since PIP5K1α was the major form in skeletal muscle, knockdown of PIP5K1α consistently inhibited myogenic differentiation while overexpression of PIP5K1α promoted differentiation and rescued the inhibitory effect of the siRNA. PIP5K1α was found to be required for AKT activation and calcium release, both of which were important for skeletal muscle differentiation. CONCLUSIONS: Taken together, these results suggest that PIP5K1α is an important regulator in myoblast differentiation.


Subject(s)
Calcium Signaling , Calcium/metabolism , Cell Differentiation , Muscle Development , Muscle, Skeletal/enzymology , Myoblasts, Skeletal/enzymology , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Animals , Cell Line , Enzyme Activation/genetics , Gene Knockdown Techniques , Mice , Muscle, Skeletal/cytology , Myoblasts, Skeletal/cytology , Myogenin/biosynthesis , Myogenin/genetics , Myosin Heavy Chains/biosynthesis , Myosin Heavy Chains/genetics , Phosphotransferases (Alcohol Group Acceptor)/genetics , Proto-Oncogene Proteins c-akt/genetics
16.
Inflammation ; 41(1): 199-212, 2018 Feb.
Article in English | MEDLINE | ID: mdl-28971270

ABSTRACT

The objective of this study is to investigate the role of Calmodulin-dependent protein kinase IV (CaMKIV) in Cardiotoxin (CTX)-induced mice muscle inflammation. CTX injection i.m. was performed to induce B6 mice acute tibialis anterior (TA) muscle injury. The mice were then injected i.p. with the recombinant CaMKIV protein or its antagonist KN-93. Immunoblotting was used to assess Calmodulin (CaM) and CaMKIV levels. Immunofluorescence was used to detect intramuscular infiltration or major histocompatibility complex (MHC)-I expression in damaged muscle. The extent of infiltration was evaluated by fluorescent intensity analysis. Cytokines/chemokines levels were determined by qPCR. CaMKIV gene knockdown in C2C12 cells was performed in order to evaluate the effects of CaMKIV on immuno-behavior of muscle cells. CTX administration induced a strong up-regulation of CaM and p-CaMKIV levels in infiltrated mononuclear cells and regenerated myofibers. In vivo adding of the recombinant CaMKIV protein enhanced intramuscular infiltration of monocytes/macrophages in damaged muscle and increased the number of proinflammatory Ly-6C+F4/80+ macrophage cells. CaMKIV protein treatment induced a striking up-regulation of mRNA levels of IL-1, IL-6, MCP-1, and MCP-3 in CD45+ cells sorted from damaged muscle; increased the infiltration of CD8+ T cells; and induced the up-regulation of MHC-I in partial regenerated myofibers, which was rarely observed in muscle damage alone. Additionally, CaMKIV protein treatment diminished the regulatory T cells (Tregs) number and led to the damaged TA muscle repair delay. In vitro CaMKIV gene knockdown reversed IFN-γ-induced up-regulation of MHC-I/II and TLR3 in the differentiated C2C12 myotubes. CaMKIV can act as an immunostimulation molecule and enhances the acute muscle inflammatory responses.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinase Type 4/metabolism , Muscle Development , Muscle, Skeletal/enzymology , Myoblasts, Skeletal/enzymology , Myositis/enzymology , Regeneration , Acute Disease , Animals , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Calcium-Calmodulin-Dependent Protein Kinase Type 4/genetics , Cell Line , Chemokines/genetics , Chemokines/metabolism , Chemotaxis, Leukocyte , Cytokines/genetics , Cytokines/metabolism , Disease Models, Animal , Gene Expression Regulation, Enzymologic , Histocompatibility Antigens Class I/metabolism , Macrophages/immunology , Macrophages/metabolism , Mice, Inbred C57BL , Monocytes/immunology , Monocytes/metabolism , Muscle, Skeletal/immunology , Muscle, Skeletal/pathology , Myoblasts, Skeletal/immunology , Myoblasts, Skeletal/pathology , Myositis/chemically induced , Myositis/genetics , Myositis/immunology , Signal Transduction , Time Factors
17.
J Nutr Biochem ; 49: 63-70, 2017 11.
Article in English | MEDLINE | ID: mdl-28886438

ABSTRACT

Ubiquitin-specific protease 19 (USP19) is a key player in the negative regulation of muscle mass during muscle atrophy. Loss-of-function approaches demonstrate that 17ß-estradiol (E2) increases USP19 expression through estrogen receptor (ER) α and consequently decreases soleus muscle mass in young female mice under physiological conditions. Daidzein is one of the main isoflavones in soy, and activates ERß-dependent transcription. Here, we investigated the effects of daidzein on E2-increased USP19 expression and E2-decreased soleus muscle mass in young female mice. Daidzein stimulated the transcriptional activity of ERß in murine C2C12 cells and down-regulated USP19 expression. Consistently, daidzein inhibited E2-induced USP19 expression in a reporter activity using a functional half-estrogen response element (hERE) from Usp19. Daidzein inhibited E2-induced recruitment of ERα and promoted recruitment of ERß to the Usp19 hERE. Dietary daidzein down-regulated the expression of USP19 at the mRNA and protein levels and increased soleus muscle mass in female mice, but not in males. In soleus muscle from ovariectomized (OVX) female mice, dietary daidzein inhibited E2-increased USP19 mRNA expression and E2-decreased muscle mass. Furthermore, E2 induced the recruitment of ERα and ERß to the hERE, whereas daidzein inhibited E2-induced recruitment of ERα, and enhanced E2-increased recruitment of ERß, to the Usp19 hERE. These results demonstrate that dietary daidzein decreases USP19 mRNA expression through ERß and increases soleus muscle mass in young female mice, but not in male mice, under physiological conditions.


Subject(s)
Dietary Supplements , Estrogen Receptor beta/agonists , Isoflavones/therapeutic use , Muscle, Skeletal/metabolism , Phytoestrogens/therapeutic use , Sarcopenia/prevention & control , Ubiquitin-Specific Proteases/antagonists & inhibitors , Active Transport, Cell Nucleus , Animals , Animals, Outbred Strains , Cell Line , Endopeptidases , Enzyme Repression , Estrogen Receptor beta/genetics , Estrogen Receptor beta/metabolism , Female , Genes, Reporter , Male , Mice , Muscle, Skeletal/enzymology , Muscle, Skeletal/pathology , Myoblasts, Skeletal/cytology , Myoblasts, Skeletal/enzymology , Myoblasts, Skeletal/metabolism , Myoblasts, Skeletal/pathology , Ovariectomy/adverse effects , Random Allocation , Response Elements , Sarcopenia/etiology , Sarcopenia/metabolism , Sarcopenia/pathology , Sex Characteristics , Signal Transduction , Ubiquitin-Specific Proteases/genetics , Ubiquitin-Specific Proteases/metabolism
18.
J Biol Chem ; 292(37): 15538-15551, 2017 09 15.
Article in English | MEDLINE | ID: mdl-28784662

ABSTRACT

Differences among fatty acids (FAs) in chain length and number of double bonds create lipid diversity. FA elongation proceeds via a four-step reaction cycle, in which the 3-hydroxyacyl-CoA dehydratases (HACDs) HACD1-4 catalyze the third step. However, the contribution of each HACD to 3-hydroxyacyl-CoA dehydratase activity in certain tissues or in different FA elongation pathways remains unclear. HACD1 is specifically expressed in muscles and is a myopathy-causative gene. Here, we generated Hacd1 KO mice and observed that these mice had reduced body and skeletal muscle weights. In skeletal muscle, HACD1 mRNA expression was by far the highest among the HACDs However, we observed only an ∼40% reduction in HACD activity and no changes in membrane lipid composition in Hacd1-KO skeletal muscle, suggesting that some HACD activities are redundant. Moreover, when expressed in yeast, both HACD1 and HACD2 participated in saturated and monounsaturated FA elongation pathways. Disruption of HACD2 in the haploid human cell line HAP1 significantly reduced FA elongation activities toward both saturated and unsaturated FAs, and HACD1 HACD2 double disruption resulted in a further reduction. Overexpressed HACD3 exhibited weak activity in saturated and monounsaturated FA elongation pathways, and no activity was detected for HACD4. We therefore conclude that HACD1 and HACD2 exhibit redundant activities in a wide range of FA elongation pathways, including those for saturated to polyunsaturated FAs, with HACD2 being the major 3-hydroxyacyl-CoA dehydratase. Our findings are important for furthering the understanding of the molecular mechanisms in FA elongation and diversity.


Subject(s)
Fatty Acids/metabolism , Hydro-Lyases/metabolism , Membrane Proteins/metabolism , Muscle, Skeletal/enzymology , Myoblasts, Skeletal/enzymology , Protein Tyrosine Phosphatases/metabolism , Animals , CRISPR-Cas Systems , Catalytic Domain , Cell Line, Tumor , Cells, Cultured , Fatty Acids/chemistry , Gene Expression Regulation, Enzymologic , Humans , Hydro-Lyases/genetics , Isoenzymes/genetics , Isoenzymes/metabolism , Male , Membrane Proteins/genetics , Mice, Knockout , Molecular Structure , Molecular Weight , Muscle, Skeletal/cytology , Muscle, Skeletal/pathology , Muscular Diseases/enzymology , Muscular Diseases/genetics , Muscular Diseases/pathology , Myoblasts, Skeletal/cytology , Myoblasts, Skeletal/pathology , Protein Tyrosine Phosphatases/genetics , Recombinant Fusion Proteins/metabolism , Substrate Specificity
19.
Mol Cell ; 67(2): 239-251.e6, 2017 Jul 20.
Article in English | MEDLINE | ID: mdl-28669802

ABSTRACT

UPF1 is an RNA helicase that orchestrates nonsense-mediated decay and other RNA surveillance pathways. While UPF1 is best known for its basal cytoprotective role in degrading aberrant RNAs, UPF1 also degrades specific, normally occurring mRNAs to regulate diverse cellular processes. Here we describe a role for UPF1 in regulated protein decay, wherein UPF1 acts as an E3 ubiquitin ligase to repress human skeletal muscle differentiation. Suppressing UPF1 accelerates myogenesis, while ectopically increasing UPF1 levels slows myogenesis. UPF1 promotes the decay of MYOD protein, a transcription factor that is a master regulator of myogenesis, while leaving MYOD mRNA stability unaffected. UPF1 acts as an E3 ligase via its RING domain to promote MYOD protein ubiquitination and degradation. Our data characterize a regulatory role for UPF1 in myogenesis, and they demonstrate that UPF1 provides a mechanistic link between the RNA and protein decay machineries in human cells.


Subject(s)
Cell Differentiation , Muscle Development , Muscle, Skeletal/enzymology , Myoblasts, Skeletal/enzymology , Trans-Activators/metabolism , Ubiquitin-Protein Ligases/metabolism , Down-Regulation , Female , HEK293 Cells , Humans , Male , Muscle, Skeletal/cytology , MyoD Protein/genetics , MyoD Protein/metabolism , Protein Domains , Proteolysis , RNA Helicases , RNA Interference , RNA Stability , RNA, Messenger/genetics , RNA, Messenger/metabolism , Signal Transduction , Time Factors , Trans-Activators/chemistry , Trans-Activators/genetics , Transcription, Genetic , Transfection , Ubiquitination
20.
Mol Cell Biol ; 37(4)2017 02 15.
Article in English | MEDLINE | ID: mdl-27920252

ABSTRACT

Skeletal myogenesis is regulated by signal transduction, but the factors and mechanisms involved are not well understood. The group I Paks Pak1 and Pak2 are related protein kinases and direct effectors of Cdc42 and Rac1. Group I Paks are ubiquitously expressed and specifically required for myoblast fusion in Drosophila We report that both Pak1 and Pak2 are activated during mammalian myoblast differentiation. One pathway of activation is initiated by N-cadherin ligation and involves the cadherin coreceptor Cdo with its downstream effector, Cdc42. Individual genetic deletion of Pak1 and Pak2 in mice has no overt effect on skeletal muscle development or regeneration. However, combined muscle-specific deletion of Pak1 and Pak2 results in reduced muscle mass and a higher proportion of myofibers with a smaller cross-sectional area. This phenotype is exacerbated after repair to acute injury. Furthermore, primary myoblasts lacking Pak1 and Pak2 display delayed expression of myogenic differentiation markers and myotube formation. These results identify Pak1 and Pak2 as redundant regulators of myoblast differentiation in vitro and in vivo and as components of the promyogenic Ncad/Cdo/Cdc42 signaling pathway.


Subject(s)
Cell Differentiation , Myoblasts, Skeletal/cytology , Myoblasts, Skeletal/enzymology , p21-Activated Kinases/metabolism , Animals , Cadherins/metabolism , Cell Adhesion Molecules/metabolism , Cell Line , Enzyme Activation , Mice , Mice, Knockout , Models, Biological , Muscle Development , NIH 3T3 Cells , Neuromuscular Junction/metabolism , Organ Size , Proto-Oncogene Proteins c-akt/metabolism , Regeneration , Signal Transduction , Synapses/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...