Next Article in Journal
Mitochondrial Dysfunction and Chronic Liver Disease
Next Article in Special Issue
Fetal Lung Cells Transfer Improves Emphysematous Change in a Mouse Model of Neutrophil Elastase-Induced Lung Emphysema
Previous Article in Journal
Antioxidant Activity of New Sulphur- and Selenium-Containing Analogues of Potassium Phenosan against H2O2-Induced Cytotoxicity in Tumour Cells
Previous Article in Special Issue
Characterization of Insulin-like Peptide (ILP) and Its Potential Role in Ovarian Development of the Cuttlefish Sepiella japonica
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

TNF-α Suppresses Apelin Receptor Expression in Mouse Quadriceps Femoris-Derived Cells

1
Department of Orthopedic Surgery, Kitasato University School of Medicine, 1-15-1 Minami-ku, Kitasato, Sagamihara City 252-0374, Kanagawa, Japan
2
Department of Biochemistry, Kitasato University School of Medicine, 1-15-1 Minami-ku, Kitasato, Sagamihara City 252-0374, Kanagawa, Japan
*
Author to whom correspondence should be addressed.
Curr. Issues Mol. Biol. 2022, 44(7), 3146-3155; https://doi.org/10.3390/cimb44070217
Submission received: 13 May 2022 / Revised: 6 July 2022 / Accepted: 8 July 2022 / Published: 8 July 2022

Abstract

:
Expression of the apelin receptor, APJ, in skeletal muscle (SM) is known to decrease with age, but the underlying mechanism remains unclear. Increased tumor necrosis factor (TNF)-α levels are observed in SM with age and are associated with muscle atrophy. To investigate the possible interconnection between TNF-α elevation and APJ reduction with aging, we investigated the effect of TNF-α on APJ expression in cells derived from the quadriceps femoris of C57BL/6J mice. Expression of Tnfa and Apj in the quadriceps femoris was compared between 4- (young) and 24-month-old (old) C57BL/6J mice (n = 10 each) using qPCR. Additionally, APJ-positive cells and TNF-α protein were analyzed by flow cytometry and Western blotting, respectively. Further, quadricep-derived cells were exposed to 0 (control) or 25 ng/mL TNF-α, and the effect on Apj expression was examined by qRT-PCR. Apj expression and the ratio of APJ-positive cells among quadricep cells were significantly lower in old compared to young mice. In contrast, levels of Tnfa mRNA and TNF-α protein were significantly elevated in old compared to young mice. Exposing young and old derived quadricep cells to TNF-α for 8 and 24 h caused Apj levels to significantly decrease. TNF-α suppresses APJ expression in muscle cells in vitro. The increase in TNF-α observed in SM with age may induce a decrease in APJ expression.

1. Introduction

Sarcopenia is a disorder that is characterized by the gradual deterioration of skeletal muscle (SM) mass and strength as a person ages, and leads to a decline in autonomy in older people [1,2]. Loss of motility and mobility is increasingly believed to be one of the most compelling indicators of poor health outcomes in older people [3,4]. Knowledge accumulated through decades of extensive research with many different animal and human models on skeletal muscle atrophy/deconditioning has provided us with a good understanding of the cellular processes implicated [5,6,7,8,9,10]. To identify other potential cellular mechanisms and improve understanding of those already discovered, is important to reveal the underlying causes of sarcopenia.
The G protein-coupled apelin receptor (APJ) is widely expressed throughout the body. APJ is implicated in various physiological processes, including energy metabolism, cardiovascular function, and angiogenesis. A recent study reported that APJ and its ligand apelin contribute to muscle metabolism and that APJ gene expression is decreased in old mice [11]. However, the cause of the decrease in APJ with age has not been fully clarified.
Interestingly, inflammatory cytokine levels are reported to be elevated in the blood and skeletal muscle tissue of sarcopenia patients [12,13,14,15,16,17,18]. Higher plasma concentrations of tumor necrosis factor (TNF)-α result in reduced muscle mass and strength in normally functioning elderly people [18]. A single intraperitoneal administration of TNF-α (100 μg/kg) to male ICR mice increased the cytoplasmic oxidative activity of muscle fibers isolated from the intercostal diaphragm and decreased the maximal force of the diaphragm [19]. A recent study reported that elevated concentrations of TNF-α in the SM of old mice are associated with atrophy [20]. Further, a strong link between TNF-α, protein turnover alteration and muscle deconditioning with aging has been reported [20,21,22,23,24,25,26]. TNF-α induced protein loss in skeletal muscle myocytes via reactive oxygen-mediated NF-κB activation [25]. TNF-α signaling induced muscle fiber-specific apoptosis [27]. Further, TNF-α upregulated Atrogin1/MAFbx, which appear to be essential for accelerated muscle protein loss [22]. Several studies have reported that APJ signaling regulates TNF-α in macrophages, adipocytes, and hepatocytes in vitro [28,29,30]. However, it remains unclear whether TNF-α regulates APJ expression in cells derived from SM.
To investigate the possible interconnection between TNF-α elevation and APJ reduction with aging, we investigated the effect of TNF-α on APJ expression in cells derived from the quadriceps femoris of C57BL/6J mice.

2. Materials and Methods

2.1. Animals

This study was conducted on male C57BL/6J mice (Jackson Laboratory Japan, Yokohama, Japan). C57BL/6J mice were housed at Jackson Laboratory Japan (Kanagawa, Japan) under a semibarrier system with controlled temperature (23 ± 2 °C), humidity (55% ± 10%), and light (12 h light/dark cycle). The study protocol was approved by the Kitasato University School of Medicine Animal Care Committee (reference number: 2021-046).
A previous study showed that muscle/body weight was reduced in 24-month-old C57BL/6 mice compare to 3-month-old C57BL/6 mice [31]. Therefore, we categorized 3-month-old mice (n = 10) as the “young” group and 24-month-old mice (n = 10) as the “old” group. Body weight (g) and muscle weight of the quadriceps femoris (mg) were measured and muscle weight (mg)/body weight (n = 10) was calculated. Apelin, Apj and Tnfa mRNA expression in the quadriceps femoris was examined using real-time PCR and compared between the two age groups. In addition, TNF-α protein expression was examined by Western blotting. To determine whether TNF-α affects apelin and APJ expression in SM, quadriceps femoris tissue from young (n = 5) and old mice (n = 5) was digested with collagenase. Muscle cells were subsequently harvested and exposed to 0 ng/mL (control: culture medium only), 2.5 ng/mL or 25 ng/mL TNF-α for 8 and 24 h. mRNA was extracted from the stimulated cells and Apelin and Apj expression was measured using real-time PCR.

2.2. Real-Time PCR

C57BL/6J mice were sacrificed by inhalation anesthesia with isoflurane. Using a scalpel, the skin and fascia of the upper leg were removed and the quadriceps femoris was harvested. The harvested tissue was then subjected to TRIzol (Invitrogen, Carlsbad, CA, USA) treatment to extract total RNA based on the manufacturer’s protocol. The total RNA formed the template for cDNA synthesis using SuperScript III RT (Thermo Fisher Scientific, CA, USA) in a PCR reaction that comprised cDNA, TB Green Premix Ex Taq (Takara, Kyoto, Japan) and a specific primer set. Primers in the primer set were fashioned on Primer Blast software and made by Hokkaido System Science Co., Ltd. (Sapporo, Japan). Table 1 lists the primer sequences adopted in this study. Amplified products were examined for specificity using melt curve analysis. Quantitative PCR was conducted on a CFX connect real-time PCR detection system (Bio-Rad, Hercules, CA, USA) with a denaturation step at 95 °C for 1 min, 40 cycles of 95 °C for 5 s and 60 °C for 30 s. We evaluated β-actin and GAPDH as housekeeping genes. Because β-actin gene differed between young and old mice, levels of each mRNA of interest were normalized to concentrations of GAPDH.

2.3. Western Blotting

Protein levels of TNF-α were measured using Western blotting. After homogenizing muscle cells in sodium dodecyl sulfate (SDS) sample buffer, the homogenates immediately heated at 95 °C for 10 min. Protein concentrations were determined by a bicinchoninic acid (BCA) assay kit (Thermo Fisher Scientific, Inc., Waltham, MA, USA). No protein degradation was confirmed in Coomassie Brilliant Blue staining. The homogenates (5 μg/lane) were subjected to SDS-polyacrylamide gel electrophoresis. The separated proteins in the gel were then electrophoretically transferred to a polyvinylidene difluoride membrane in blotting buffer. The membrane was subsequently treated with 10% skim milk in TBST for 30 min at 25 °C to prevent non-specific reactions before incubating with anti-TNF-α antibody (1:1000; catalog number. Ab6671, Abcam Cambridge, UK) or anti-GAPDH antibody (1:5000; FUJIFILM Wako Pure Chemical Co., Osaka, Japan) for 60 min at 25 °C. After further incubating with goat anti-rabbit antibody conjugated to HRP (catalog number. 211-035-109, RRID: AB_2339150, Jackson Immuno Research Laboratories; West Grove, PA, USA) for 60 min at 25 °C, the membrane was washed a final time. Protein bands were subsequently visualized using enhanced chemiluminescence (catalog number 07880, Chemi-Lumi One L; Nacalai Tesque, Kyoto, Japan) and a luminescent image analyzer with a CCD imager (LAS-4000mini; Fuji Photo Film Co., Tokyo, Japan). Relative TNF-α expression was normalized to GAPDH.

2.4. Flow Cytometry

Tissue samples of the quadriceps femoris taken from young and old mice (n = 5 each) were treated with a 20 mL solution of 0.1% collagenase (Catalog Number. 03222364, Fujifilm Wako Pure Chemical Corporation, Osaka, Japan) at 37 °C for 1 h. The digested samples were then filtered through a nylon mesh filter (pluriStrainer 100 µm, pluriSelect, Leipzig, Germany) to obtain cell suspensions. The cells were then treated with the following antibodies: anti-CD45-PE/Cy7 (Clone: 30-F11, Catalog number 103113, BioLegend, CA, USA) and anti-Sca1-APC-Cy7 (Clone: D7, Catalog number 108126, BioLegend) for 1 h at 4 °C. CD45 is a marker for pan-hematopoietic cells and Sca1 is a marker for mature myocytes. After further treatment with a fixation/permeabilization solution (catalog number 420801, BioLegend), the cells were exposed to FITC-conjugated anti-APJ antibody, prepared using an FITC conjugation kit (Lightning-Link conjugation kit, Abcam) and unlabeled anti-APJ antibody (Cat. No. 20341-1-AP, Proteintech, CA, USA) for 30 min at 4 °C. After washing in wash buffer twice, the labeled cells were used for flow cytometry. The procedure involved acquiring 50,000 total events using a BD FACSVerse system (BD Biosciences, San Jose, CA, USA) and analysis of the findings using FlowJo v10.7 (Tree Star, Ashland, OR, USA). Negative gates were set based on the isotype control.

2.5. Muscle Cell Culture

To extract mononuclear cells from the quadriceps femoris of young and old mice (n = 5), tissue samples were treated with a 20 mL solution of 0.1% type I collagenase for 60 min at 37 °C. The harvested cells (1 × 104 cells/cm2) containing heterogenous populations were cultured in α-minimal essential media (Gibco Life Technologies, Carlsbad, CA, USA) + 10% fetal bovine serum (Gibco Life Technologies; lot no. 42Q0170K) in six-well plates for 7 days. The cultured cells were then exposed to 0 ng/mL (control: culture medium only), 2.5 ng/mL mouse TNF-α, or 25 ng/mL TNF-α for 8 or 24 h. The gene expression of Apelin and Apj in muscle-derived cells was determined using real-time PCR in the same manner as that described above.

2.6. Statistics

Statistical analyses were performed using SPSS version 28.0.0.0 (190) (IBM, Armonk, NY, USA). The Shapiro–Wilk test was used to test for normality, and Levene’s test for the homogeneity of the variance. Mann–Whitney U tests were used to compare body weight and muscle mass between the two age groups. The unpaired t-test was used to compare muscle mass/body weight between the two age groups. As the gene and protein expression data were not normally distributed, the Mann–Whitney U test was used to compare gene and protein expression between the two age groups. Two-way ANOVA with the Bonferroni post-hoc test was used to compare the gene expression among control, 2.5 ng/mL TNF-α-, and 25 ng/mL TNF-α-stimulated cells. p < 0.05 was considered significant. All values are expressed as the mean ± standard deviation (SD).

3. Results

3.1. Muscle Mass of the Quadriceps of Young and Old Mice

Body weight was significantly higher in old mice (32.6 ± 2.9 g) than in young mice (24.5 ± 1.3 g, p < 0.001; Figure 1A). Muscle mass did not significantly differ between young (186.5 ± 16.1 mg) and old mice (191.9 ± 45.0 mg) (p = 0.481; Figure 1B). However, muscle mass/body weight was significantly lower in old mice (5.8 ± 1.0 mg/g) than in young mice (7.6 ± 0.6 mg/g, p < 0.001; Figure 1C).

3.2. Expression of Apelin and APJ in the Quadriceps of Young and Old Mice

Apelin expression was not significantly different (1.00 ± 0.12 (young) vs. 0.79 ± 0.06 (old), p = 0.253, Figure 2A), whereas Apj was significantly decreased in old compared to young mice (1.00 ± 0.06 (young) vs.0.38 ± 0.06 (Old), p < 0.001, Figure 2B). Further, the ratio of APJ-positive cells was also reduced in old mice among both Sca1-positive and Sca1-negative cells (Sca1-positive: 1.26 ± 0.50 (young) vs. 0.08 ± 0.04 (old), p = 0.016, Sca1-negative: 0.72 ± 0.22 (young) vs. 0.11 ± 0.02 (old), p = 0.016, Figure 3A,B).

3.3. Expression of TNF-α in the Quadriceps of Young and Old Mice

mRNA expression of Tnfa was significantly elevated in old compared to young mice (1.00 ± 0.26 (young) vs. 2.47 ± 0.52 (old), p = 0.034, Figure 4A). Western blotting showed that protein expression of TNF-α was likewise increased in old mice (1.00 ± 0.09 (young) vs. 2.52 ± 0.26 (old), p = 0.006, Figure 4B).

3.4. Effect of TNF-α on Apj Expression

As our findings indicated that the expression level of APJ differed between young and old mice, we next evaluated whether the response of muscle cells to TNF-α was also different between the two age groups. Relative Apelin mRNA expression in young mice-derived cells is shown in Figure 5A, namely, control (8 h, 1.00 ± 0.07; 24 h, 1.00 ± 0.08), 2.5 ng/mL TNF-α (8 h, 1.64 ± 0.06; 24 h, 0.93 ± 0.01), and 25 ng/mL TNF-α (8 h, 1.64 ± 0.07; 24 h, 1.31 ± 0.01).
TNF-α significantly increased Apelin mRNA expression in young mice-derived muscle cells for 8 h (control vs. 2.5 ng/mL TNF-α: p < 0.001, control vs. 25 ng/mL TNF-α: p < 0.001, Figure 4A) and for 24 h (control vs. 25 ng/mL TNF-α: p < 0.001; Figure 5A).
Relative Apelin mRNA expression in old mice-derived cells is shown in Figure 5B, namely, control (8 h, 1.00 ± 0.21, 24 h, 1.00 ± 0.22), 2.5 ng/mL TNF-α (8 h, 1.37 ± 0.20; 24 h, 1.21 ± 0.17), and 25 ng/mL TNF-α (8 h, 1.24 ± 0.20; 24 h, 1.05 ± 0.18). No significant increase was observed in old mice-derived muscle cells (Figure 5B).
Relative Apj mRNA expression in young mice-derived cells is shown in Figure 5C, as control (8 h, 1.00 ± 0.07; 24 h, 1.00 ± 0.08), 2.5 ng/mL TNF-α (8 h, 0.59 ± 0.06; 24 h, 0.57 ± 0.05), and 25 ng/mL TNF-α (8 h, 0.42 ± 0.01; 24 h, 0.39 ± 0.03). Relative Apj mRNA expression in old mice-derived cells is shown in Figure 5D, as control (8 h, 1.00 ± 0.19, 24 h, 1.00 ± 0.20), 2.5 ng/mL TNF-α (8 h, 0.35 ± 0.06; 24 h, 0.60 ± 0.07), and 25 ng/mL TNF-α (8 h, 0.17 ± 002; 24 h, 0.18 ± 0.03). Apj was significantly reduced in both young and old mice-derived muscle cells following stimulation with exogenous TNF-α for 8 h (young, control vs. 25 ng/mL TNF-α, p < 0.001; old, control vs. 2.5 ng/mL TNF-α: p = 0.012, control vs. 25 ng/mL TNF-α: p = 0.009, Figure 4B) and 24 h (young, control vs. 25 ng/mL TNF-α, p < 0.001; old, control vs. 25 ng/mL TNF-α: p = 0.007, Figure 5C,D).

4. Discussion

The purpose of this study was to examine interconnection between TNF-α elevation and APJ reduction with aging. We showed that old mice had reduced Apj expression and a reduced ratio of APJ-positive cells compared to young mice. In contrast, they had significantly higher concentrations of TNF-α than young mice. Further, exposing muscle-derived cells to exogenous TNF-α caused Apj mRNA expression to significantly decrease. Together, our results suggest that the reduction in APJ in old mice may be associated with increased TNF-α.
Sarcopenia has been extensively studied using mouse models. Mice have a lifespan of 2–3 years [32]. The ratio of muscle/body weight has been proposed to be a useful sarcopenia index in rodent [33]. A previous study reported that loss of muscle mass (muscle weight/body weight) first becomes evident in 24-month-old C57BL/6J mice [31]. Consistent with a previous study [31], a lower muscle weight/body weight was observed in old mice (24-month-old) compared to young mice. Therefore, we used 24-month-old mice as an aged model. Sarcopenia is defined by low levels of measures for three parameters: (1) muscle strength, (2) muscle quantity/quality and (3) physical performance as an indicator of severity [6]. In our study, we did not assess muscle strength, quality, or physical performance. In addition, muscle mass did not differ between young and old mice. Therefore, the mice used in this study may be insufficient as a sarcopenia model.
APJ has been previously reported to be associated with age-related muscle atrophy [11]. Pax7-expressing muscle stem cells express APJ, and the number these cells is reduced with age. In contrast, apelin stimulates glucose uptake and Akt phosphorylation in myotubes, suggesting that mature myogenic cells also express APJ. Previous studies have implicated Sca1 as a regulator of differentiation in myogenic cells [34]. While myoblasts are negative for Sca1, mRNA expression increases upon myogenic differentiation. In our study, we observed APJ-positive cells among both Sca1-positive and Sca1-negative cells, and that their ratio decreased in old mice. Our results thus suggest that reduced APJ expression with age reflects decreased expression in both immature and mature myogenic populations.
Several studies have shown that TNF-α rises with age in mice and humans [20,27,35]. Plasma TNF-α protein level increases with age [27]. TNF-α mRNA and protein levels are elevated in the SM of frail elderly compared to healthy young men and women [35]. Real-time PCR and flow cytometric analysis has shown that TNF-α mRNA expression in immune cells and TNF-α protein-positive macrophages are increased in skeletal muscle of old mice [20]. Similar to a previous report [20], we confirmed that TNF-α mRNA and protein expression in SM was significantly elevated in old compared to young mice. Stimulation with TNF-α significantly reduced APJ expression in muscle-derived cells from young and old mice compared to vehicle control cells. The reduced APJ expression in old mice may be associated with elevated TNF-α levels.
A previous study reported that TNF-α stimulated apelin expression in mice and human adipose tissue [29]. Consistent with this report [29], TNF-α also stimulated Apelin mRNA expression in young mice-derived muscle cells. However, TNF-α failed to stimulate apelin expression in old mice-derived muscle cells. Apelin ameliorates TNF-α mediated physiological changes in hepatocytes [28], suggesting that apelin exhibits an anti-inflammatory role toward TNF-α-induced inflammation. Lack of negative feedback by apelin may result in an elevation of inflammatory state by the TNF-a of muscle in old mice. Differences in the response of young and aged cells to inflammatory stimuli have been reported [36,37]. For example, adipocytes from old mice produce more IL-6, TNF-α, and PGE2 than those from young mice [37]. In addition, differentiation conditions could also alter cytokine response [38]. The muscle-derived used cells in the present study represented a heterogenous population and the proportion of differentiated/undifferentiated cells differed between young and old mice cells. Changes in cell phenotype with aging or a different proportion of cell populations may be associated with the different response to TNF-α in apelin expression between young and old-derived muscle cells.
There were several limitations in this study. First, only two time points were investigated. To better understand the pathogenesis of sarcopenia, further studies should analyze changes across a greater number of time points. Second, we only used muscle mass as an indicator of the pathogenesis of sarcopenia and were unable to examine the pathology of the tissue. Finally, it remains unclear whether TNF-α directly regulates APJ expression.

5. Conclusions

TNF-α suppresses APJ expression in muscle cells in vitro. The increase in TNF-α observed in SM with age may induce a decrease in APJ expression.

Author Contributions

T.K., K.U. and G.I. designed the experiments. T.K., K.U., M.I., M.M., R.T., K.F., Y.O. and A.T. collected the data. T.K., K.U., M.I. and Y.O. performed data analysis and interpretation. T.K., K.U., G.I. and M.T. wrote the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

There was no funding source for this study.

Institutional Review Board Statement

All experimental protocols in this animal study were reviewed and approved by the Kitasato University School of Medicine Animal Care Committee (2021-046).

Informed Consent Statement

Not applicable.

Data Availability Statement

The data presented in this study are available on request from the corresponding author.

Acknowledgments

We thank Akari Kobayashi, Ayumi Mineo, Yuta Nanri, Daisuke Ishii, Maho Tsuchiya, Hiroiki Saito, and Motoki Makabe for their helpful support during the study.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Anker, S.D.; Morley, J.E.; von Haehling, S. Welcome to the ICD-10 code for sarcopenia. J. Cachexia Sarcopenia Muscle 2016, 7, 512–514. [Google Scholar] [CrossRef] [PubMed]
  2. Cruz-Jentoft, A.J.; Baeyens, J.P.; Bauer, J.M.; Boirie, Y.; Cederholm, T.; Landi, F.; Martin, F.C.; Michel, J.P.; Rolland, Y.; Schneider, S.M.; et al. Sarcopenia: European consensus on definition and diagnosis: Report of the European Working Group on Sarcopenia in Older People. Age Ageing 2010, 39, 412–423. [Google Scholar] [CrossRef] [Green Version]
  3. Filippin, L.I.; Teixeira, V.N.; da Silva, M.P.; Miraglia, F.; da Silva, F.S. Sarcopenia: A predictor of mortality and the need for early diagnosis and intervention. Aging Clin. Exp. Res. 2015, 27, 249–254. [Google Scholar] [CrossRef] [PubMed]
  4. Murphy, R.A.; Ip, E.H.; Zhang, Q.; Boudreau, R.M.; Cawthon, P.M.; Newman, A.B.; Tylavsky, F.A.; Visser, M.; Goodpaster, B.H.; Harris, T.B.; et al. Transition to sarcopenia and determinants of transitions in older adults: A population-based study. J. Gerontol. A Biol. Sci. Med. Sci. 2014, 69, 751–758. [Google Scholar] [CrossRef] [PubMed]
  5. Christian, C.J.; Benian, G.M. Animal models of sarcopenia. Aging Cell 2020, 19, e13223. [Google Scholar] [CrossRef]
  6. Cruz-Jentoft, A.J.; Bahat, G.; Bauer, J.; Boirie, Y.; Bruyere, O.; Cederholm, T.; Cooper, C.; Landi, F.; Rolland, Y.; Sayer, A.A.; et al. Sarcopenia: Revised European consensus on definition and diagnosis. Age Ageing 2019, 48, 16–31. [Google Scholar] [CrossRef] [Green Version]
  7. Doherty, T.J. Invited review: Aging and sarcopenia. J. Appl. Physiol. (1985) 2003, 95, 1717–1727. [Google Scholar] [CrossRef] [Green Version]
  8. Marzetti, E.; Calvani, R.; Tosato, M.; Cesari, M.; Di Bari, M.; Cherubini, A.; Collamati, A.; D’Angelo, E.; Pahor, M.; Bernabei, R.; et al. Sarcopenia: An overview. Aging Clin. Exp. Res. 2017, 29, 11–17. [Google Scholar] [CrossRef]
  9. Tournadre, A.; Vial, G.; Capel, F.; Soubrier, M.; Boirie, Y. Sarcopenia. Jt. Bone Spine 2019, 86, 309–314. [Google Scholar] [CrossRef]
  10. Xie, W.Q.; He, M.; Yu, D.J.; Wu, Y.X.; Wang, X.H.; Lv, S.; Xiao, W.F.; Li, Y.S. Mouse models of sarcopenia: Classification and evaluation. J. Cachexia Sarcopenia Muscle 2021, 12, 538–554. [Google Scholar] [CrossRef]
  11. Vinel, C.; Lukjanenko, L.; Batut, A.; Deleruyelle, S.; Pradere, J.P.; Le Gonidec, S.; Dortignac, A.; Geoffre, N.; Pereira, O.; Karaz, S.; et al. The exerkine apelin reverses age-associated sarcopenia. Nat. Med. 2018, 24, 1360–1371. [Google Scholar] [CrossRef] [PubMed]
  12. Bian, A.L.; Hu, H.Y.; Rong, Y.D.; Wang, J.; Wang, J.X.; Zhou, X.Z. A study on relationship between elderly sarcopenia and inflammatory factors IL-6 and TNF-alpha. Eur. J. Med. Res. 2017, 22, 25. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Cesari, M.; Kritchevsky, S.B.; Baumgartner, R.N.; Atkinson, H.H.; Penninx, B.W.; Lenchik, L.; Palla, S.L.; Ambrosius, W.T.; Tracy, R.P.; Pahor, M. Sarcopenia, obesity, and inflammation--results from the Trial of Angiotensin Converting Enzyme Inhibition and Novel Cardiovascular Risk Factors study. Am. J. Clin. Nutr. 2005, 82, 428–434. [Google Scholar] [CrossRef]
  14. Cohen, T.V.; Many, G.M.; Fleming, B.D.; Gnocchi, V.F.; Ghimbovschi, S.; Mosser, D.M.; Hoffman, E.P.; Partridge, T.A. Upregulated IL-1beta in dysferlin-deficient muscle attenuates regeneration by blunting the response to pro-inflammatory macrophages. Skelet. Muscle 2015, 5, 24. [Google Scholar] [CrossRef] [Green Version]
  15. De Almeida, P.; Tomazoni, S.S.; Frigo, L.; de Carvalho Pde, T.; Vanin, A.A.; Santos, L.A.; Albuquerque-Pontes, G.M.; De Marchi, T.; Tairova, O.; Marcos, R.L.; et al. What is the best treatment to decrease pro-inflammatory cytokine release in acute skeletal muscle injury induced by trauma in rats: Low-level laser therapy, diclofenac, or cryotherapy? Lasers Med. Sci. 2014, 29, 653–658. [Google Scholar] [CrossRef] [PubMed]
  16. Krabbe, K.S.; Pedersen, M.; Bruunsgaard, H. Inflammatory mediators in the elderly. Exp. Gerontol. 2004, 39, 687–699. [Google Scholar] [CrossRef]
  17. Schrager, M.A.; Metter, E.J.; Simonsick, E.; Ble, A.; Bandinelli, S.; Lauretani, F.; Ferrucci, L. Sarcopenic obesity and inflammation in the InCHIANTI study. J. Appl. Physiol. (1985) 2007, 102, 919–925. [Google Scholar] [CrossRef] [PubMed]
  18. Visser, M.; Pahor, M.; Taaffe, D.R.; Goodpaster, B.H.; Simonsick, E.M.; Newman, A.B.; Nevitt, M.; Harris, T.B. Relationship of interleukin-6 and tumor necrosis factor-alpha with muscle mass and muscle strength in elderly men and women: The Health ABC Study. J. Gerontol. A Biol. Sci. Med. Sci. 2002, 57, M326–M332. [Google Scholar] [CrossRef] [Green Version]
  19. Hardin, B.J.; Campbell, K.S.; Smith, J.D.; Arbogast, S.; Smith, J.; Moylan, J.S.; Reid, M.B. TNF-alpha acts via TNFR1 and muscle-derived oxidants to depress myofibrillar force in murine skeletal muscle. J. Appl. Physiol. (1985) 2008, 104, 694–699. [Google Scholar] [CrossRef]
  20. Li, J.; Yi, X.; Yao, Z.; Chakkalakal, J.V.; Xing, L.; Boyce, B.F. TNF Receptor-Associated Factor 6 Mediates TNFalpha-Induced Skeletal Muscle Atrophy in Mice During Aging. J. Bone Miner. Res. 2020, 35, 1535–1548. [Google Scholar] [CrossRef]
  21. Lang, C.H.; Frost, R.A.; Nairn, A.C.; MacLean, D.A.; Vary, T.C. TNF-alpha impairs heart and skeletal muscle protein synthesis by altering translation initiation. Am. J. Physiol. Endocrinol. Metab. 2002, 282, E336–E347. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Li, Y.P.; Chen, Y.; John, J.; Moylan, J.; Jin, B.; Mann, D.L.; Reid, M.B. TNF-alpha acts via p38 MAPK to stimulate expression of the ubiquitin ligase atrogin1/MAFbx in skeletal muscle. FASEB J. 2005, 19, 362–370. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Li, Y.P.; Lecker, S.H.; Chen, Y.; Waddell, I.D.; Goldberg, A.L.; Reid, M.B. TNF-alpha increases ubiquitin-conjugating activity in skeletal muscle by up-regulating UbcH2/E220k. FASEB J. 2003, 17, 1048–1057. [Google Scholar] [CrossRef]
  24. Li, Y.P.; Reid, M.B. NF-kappaB mediates the protein loss induced by TNF-alpha in differentiated skeletal muscle myotubes. Am. J. Physiol. Regul. Integr. Comp. Physiol. 2000, 279, R1165–R1170. [Google Scholar] [CrossRef] [PubMed]
  25. Li, Y.P.; Schwartz, R.J.; Waddell, I.D.; Holloway, B.R.; Reid, M.B. Skeletal muscle myocytes undergo protein loss and reactive oxygen-mediated NF-kappaB activation in response to tumor necrosis factor alpha. FASEB J. 1998, 12, 871–880. [Google Scholar]
  26. Reid, M.B.; Li, Y.P. Tumor necrosis factor-alpha and muscle wasting: A cellular perspective. Respir. Res. 2001, 2, 269–272. [Google Scholar] [CrossRef]
  27. Phillips, T.; Leeuwenburgh, C. Muscle fiber specific apoptosis and TNF-alpha signaling in sarcopenia are attenuated by life-long calorie restriction. FASEB J. 2005, 19, 668–670. [Google Scholar] [CrossRef]
  28. Chu, J.; Zhang, H.; Huang, X.; Lin, Y.; Shen, T.; Chen, B.; Man, Y.; Wang, S.; Li, J. Apelin ameliorates TNF-alpha-induced reduction of glycogen synthesis in the hepatocytes through G protein-coupled receptor APJ. PLoS ONE 2013, 8, e57231. [Google Scholar] [CrossRef]
  29. Daviaud, D.; Boucher, J.; Gesta, S.; Dray, C.; Guigne, C.; Quilliot, D.; Ayav, A.; Ziegler, O.; Carpene, C.; Saulnier-Blache, J.S.; et al. TNFalpha up-regulates apelin expression in human and mouse adipose tissue. FASEB J. 2006, 20, 1528–1530. [Google Scholar] [CrossRef]
  30. Zhang, X.; Ye, Q.; Gong, D.; Lv, Y.; Cheng, H.; Huang, C.; Chen, L.; Zhao, Z.; Li, L.; Wei, X.; et al. Apelin-13 inhibits lipoprotein lipase expression via the APJ/PKCalpha/miR-361–5p signaling pathway in THP-1 macrophage-derived foam cells. Acta Biochim. et Biophys. Sin. 2017, 49, 530–540. [Google Scholar] [CrossRef] [Green Version]
  31. Shavlakadze, T.; McGeachie, J.; Grounds, M.D. Delayed but excellent myogenic stem cell response of regenerating geriatric skeletal muscles in mice. Biogerontology 2010, 11, 363–376. [Google Scholar] [CrossRef] [PubMed]
  32. Peto, R.; Roe, F.J.; Lee, P.N.; Levy, L.; Clack, J. Cancer and ageing in mice and men. Br. J. Cancer 1975, 32, 411–426. [Google Scholar] [CrossRef] [PubMed]
  33. Edstrom, E.; Ulfhake, B. Sarcopenia is not due to lack of regenerative drive in senescent skeletal muscle. Aging Cell 2005, 4, 65–77. [Google Scholar] [CrossRef] [PubMed]
  34. Mitchell, P.O.; Mills, T.; O’Connor, R.S.; Kline, E.R.; Graubert, T.; Dzierzak, E.; Pavlath, G.K. Sca-1 negatively regulates proliferation and differentiation of muscle cells. Dev. Biol. 2005, 283, 240–252. [Google Scholar] [CrossRef] [PubMed]
  35. Greiwe, J.S.; Cheng, B.; Rubin, D.C.; Yarasheski, K.E.; Semenkovich, C.F. Resistance exercise decreases skeletal muscle tumor necrosis factor alpha in frail elderly humans. FASEB J. 2001, 15, 475–482. [Google Scholar] [CrossRef] [PubMed]
  36. Solana, R.; Tarazona, R.; Gayoso, I.; Lesur, O.; Dupuis, G.; Fulop, T. Innate immunosenescence: Effect of aging on cells and receptors of the innate immune system in humans. Semin. Immunol. 2012, 24, 331–341. [Google Scholar] [CrossRef]
  37. Wu, D.; Ren, Z.; Pae, M.; Guo, W.; Cui, X.; Merrill, A.H.; Meydani, S.N. Aging up-regulates expression of inflammatory mediators in mouse adipose tissue. J. Immunol. 2007, 179, 4829–4839. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  38. Uchida, K.; Urabe, K.; Naruse, K.; Ujihira, M.; Mabuchi, K.; Itoman, M. Comparison of the cytokine-induced migratory response between primary and subcultured populations of rat mesenchymal bone marrow cells. J. Orthop. Sci. 2007, 12, 484–492. [Google Scholar] [CrossRef]
Figure 1. Muscle mass of the quadriceps of young and old mice. (A) Body weight (g), (B) muscle mass (mg), and (C) muscle mass (mg)/body weight (g) in young (3-month-old) and old (24-month-old) mice. Body weight was significantly higher in old mice than in young mice (p < 0.001; (A)). Muscle mass did not significantly differ between young and old mice (p = 0.481; (B)). However, muscle mass/body weight was significantly lower in old mice than in young mice (p < 0.001; (C)). Data are expressed as the mean ± standard deviation (SD). Asterisks indicate p < 0.05.
Figure 1. Muscle mass of the quadriceps of young and old mice. (A) Body weight (g), (B) muscle mass (mg), and (C) muscle mass (mg)/body weight (g) in young (3-month-old) and old (24-month-old) mice. Body weight was significantly higher in old mice than in young mice (p < 0.001; (A)). Muscle mass did not significantly differ between young and old mice (p = 0.481; (B)). However, muscle mass/body weight was significantly lower in old mice than in young mice (p < 0.001; (C)). Data are expressed as the mean ± standard deviation (SD). Asterisks indicate p < 0.05.
Cimb 44 00217 g001
Figure 2. Relative expression of Apelin, and Apj mRNA in the quadriceps femoris. (A) Apelin and (B) APJ expression in young (3-month-old) and old (24-month-old) mice. There was no difference in Apelin mRNA expression between young and old mice (Figure 1A). Apj mRNA expression was significantly lower in old mice than in young mice (p < 0.001, Figure 1B). Data are expressed as the mean ± standard deviation (SD). Asterisks indicate p < 0.05.
Figure 2. Relative expression of Apelin, and Apj mRNA in the quadriceps femoris. (A) Apelin and (B) APJ expression in young (3-month-old) and old (24-month-old) mice. There was no difference in Apelin mRNA expression between young and old mice (Figure 1A). Apj mRNA expression was significantly lower in old mice than in young mice (p < 0.001, Figure 1B). Data are expressed as the mean ± standard deviation (SD). Asterisks indicate p < 0.05.
Cimb 44 00217 g002
Figure 3. APJ−positive cells in the quadriceps femoris. (A,B) Dot plot analysis of CD45-negative myogenic cells. X-axis, Sca-1; Y-axis, APJ. (C,D) Ratio of APJ-positive cells among CD45-negative/Sca1-negative cells © and CD45-negative/Sca1-positive cells (D) in young (3-month-old) and old (24-month-old) mice. Significant reduction in APJ-positive cells was found in Sca1+ (p = 0.016) and Sca1− cells (p = 0.016). Data are expressed as the mean ± standard deviation (SD). Asterisks indicate p < 0.05 (Mann–Whitney U test).
Figure 3. APJ−positive cells in the quadriceps femoris. (A,B) Dot plot analysis of CD45-negative myogenic cells. X-axis, Sca-1; Y-axis, APJ. (C,D) Ratio of APJ-positive cells among CD45-negative/Sca1-negative cells © and CD45-negative/Sca1-positive cells (D) in young (3-month-old) and old (24-month-old) mice. Significant reduction in APJ-positive cells was found in Sca1+ (p = 0.016) and Sca1− cells (p = 0.016). Data are expressed as the mean ± standard deviation (SD). Asterisks indicate p < 0.05 (Mann–Whitney U test).
Cimb 44 00217 g003
Figure 4. Tnfa mRNA and TNF-α protein expression in the quadriceps femoris. (A) Tnfa mRNA levels in young (3-month-old) and old (24-month-old) mice. (B) Image of a Western blot showing TNF-α protein expression relative to GAPDH at three (3M) and 24 months (24M). (C) TNF-α protein levels in the quadriceps femoris of young (3-month-old) and old (24-month-old) mice. Significant elevation of TNF-α mRNA and protein levels was found in the old group. Data are expressed as the mean ± standard deviation (SD). Asterisks indicate p < 0.05.
Figure 4. Tnfa mRNA and TNF-α protein expression in the quadriceps femoris. (A) Tnfa mRNA levels in young (3-month-old) and old (24-month-old) mice. (B) Image of a Western blot showing TNF-α protein expression relative to GAPDH at three (3M) and 24 months (24M). (C) TNF-α protein levels in the quadriceps femoris of young (3-month-old) and old (24-month-old) mice. Significant elevation of TNF-α mRNA and protein levels was found in the old group. Data are expressed as the mean ± standard deviation (SD). Asterisks indicate p < 0.05.
Cimb 44 00217 g004
Figure 5. Effect of TNF-α on Apelin and Apj expression. Relative expression of Apelin (A,B) and Apj (C,D) in young and old mice-derived muscle cells after stimulation with 0 (control), 2.5, or 25 ng/mL mouse recombinant TNF-α for 8 and 24 h. Significant elevation of Apelin mRNA expression was only observed in young mice-derived muscle cells after TNF-α stimulation for 8 and 24 h (A,B). Apj mRNA expression was significantly reduced following TNF-α stimulation for 8 and 24 h in both young and old mice-derived muscle cells (C,D). Data are expressed as the mean ± standard deviation (SD). Asterisks indicate p < 0.05.
Figure 5. Effect of TNF-α on Apelin and Apj expression. Relative expression of Apelin (A,B) and Apj (C,D) in young and old mice-derived muscle cells after stimulation with 0 (control), 2.5, or 25 ng/mL mouse recombinant TNF-α for 8 and 24 h. Significant elevation of Apelin mRNA expression was only observed in young mice-derived muscle cells after TNF-α stimulation for 8 and 24 h (A,B). Apj mRNA expression was significantly reduced following TNF-α stimulation for 8 and 24 h in both young and old mice-derived muscle cells (C,D). Data are expressed as the mean ± standard deviation (SD). Asterisks indicate p < 0.05.
Cimb 44 00217 g005
Table 1. Primer sequences.
Table 1. Primer sequences.
GeneDirectionPrimer Sequence (5′–3′)Product Size (bp)
ApelinFTGA ATC TGA GGC TCT GCG TG223
RATG GGG CCC TTA TGG GAG AG
ApjFTAC GCC AGT GTC TTT TGC CT159
RCAC CAT GAC AGG CAC AGC TA
TnfaFCTG AAC TTC GGG GTG ATC GG122
RGGC TTG TCA CTC GAA TTT TGA GA
GAPDHFAAC TTT GGC ATT GTG GAA GG223
RACA CATT GGG GGT AGG AAC A
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Koyama, T.; Uchida, K.; Itakura, M.; Miyagi, M.; Tazawa, R.; Inoue, G.; Fukushima, K.; Ohashi, Y.; Tsukada, A.; Takaso, M. TNF-α Suppresses Apelin Receptor Expression in Mouse Quadriceps Femoris-Derived Cells. Curr. Issues Mol. Biol. 2022, 44, 3146-3155. https://doi.org/10.3390/cimb44070217

AMA Style

Koyama T, Uchida K, Itakura M, Miyagi M, Tazawa R, Inoue G, Fukushima K, Ohashi Y, Tsukada A, Takaso M. TNF-α Suppresses Apelin Receptor Expression in Mouse Quadriceps Femoris-Derived Cells. Current Issues in Molecular Biology. 2022; 44(7):3146-3155. https://doi.org/10.3390/cimb44070217

Chicago/Turabian Style

Koyama, Tomohisa, Kentaro Uchida, Makoto Itakura, Masayuki Miyagi, Ryo Tazawa, Gen Inoue, Kensuke Fukushima, Yoshihisa Ohashi, Ayumi Tsukada, and Masashi Takaso. 2022. "TNF-α Suppresses Apelin Receptor Expression in Mouse Quadriceps Femoris-Derived Cells" Current Issues in Molecular Biology 44, no. 7: 3146-3155. https://doi.org/10.3390/cimb44070217

Article Metrics

Back to TopTop