Background/Aims: Poor viability of transplanted mesenchymal stem cells (MSCs) within the ischemic heart limits their therapeutic potential for cardiac repair. Globular adiponectin (gAPN) exerts anti-apoptotic effects on several types of stem cells. Herein, we investigated the effect of gAPN on the MSCs against apoptosis induced by hypoxia and serum deprivation (H/SD). Methods: MSCs exposed to H/SD conditions were treated with different concentrations of gAPN. To identify the main type of receptor, MSCs were transfected with siRNA targeting adiponectin receptor 1 or 2 (AdipoR1 or AdipoR2). To elucidate the downstream pathway, MSCs were pre-incubated with AMPK inhibitor Compound C. Apoptosis, caspase-3 activity and mitochondrial membrane potential were evaluated. Results: H/SD-induced MSCs apoptosis and caspase-3 activation were attenuated by gAPN in a concentration-dependent manner. gAPN increased Bcl-2 and decreased Bax expressions. The loss of mitochondrial membrane potential induced by H/SD was also abolished by gAPN. The protective effect of gAPN was significantly attenuated after the knockdown of AdipoR1 rather than AdipoR2. Moreover, Compound C partly suppressed the anti-apoptotic effect of gAPN. Conclusions: gAPN inhibits H/SD-induced apoptosis in MSCs via AdipoR1-mediated pathway, possibly linked to the activation of AMPK. gAPN may be a novel survival factor for MSCs in the ischemic engraftment environment.

Stem cell therapy is a promising new approach for the treatment of ischemic heart diseases [1]. Among all types of cells under investigation, mesenchymal stem cells (MSCs) are most widely studied due to the abundance of autologous sources and the ability to repair the injured myocardium [2,3,4]. Clinical trials revealed that injection of MSCs favorably affected the functional capacity, quality of life, and ventricular remodeling in patients with ischemic cardiomyopathy [5]. However, data indicated that there appeared to be slight improvement of left ventricular ejection fraction after MSCs transplantation for acute myocardial infarction [6,7]. This was probably attributed to the low survival rate of MSCs after being exposed to the hostile engraftment environment with various proapoptotic or cytoxic factors [8], including ischemia, oxidative stress, inflammation and fibrosis [9]. It was demonstrated that hypoxia and serum deprivation (H/SD), combining the two components of ischemic injury, induced programmed MSCs death through the mitochondrial apoptotic pathway [10]. Therefore, identifying factors that prevent this process and enhance the viability of MSCs under the harsh microenvironment in the ischemic heart could be crucial for MSCs' successful utilization in cellular therapy.

Adiponectin (APN, also known as Acrp30) is an adipocytokine predominantly expressed in adipocytes. It exists as two forms: the full-length peptide (fAPN) and the globular C-terminal domain (gAPN). As a product of proteolytic cleavage, gAPN is biologically active and more potent in physiological actions [11,12]. Apart from its well-characterized role in fat tissue metabolism and insulin resistance [13], accumulating evidences established the cardioprotective properties of gAPN [14,15,16,17]. Much of this beneficial effect was attributed to the anti-apoptotic actions of gAPN on cardiovascular cells [18,19,20]. Besides, previous experiments revealed that gAPN played an important modulatory role on the survival and proliferation of several types of stem cells [21,22,23,24,25,26]. More importantly, gAPN has recently been found to promote the proliferation and migration of MSCs [27]. Taken the anti-apoptotic and stem cell-regulatory properties together, it is reasonable to hypothesize that gAPN acts as an anti-apoptotic factor in MSCs under H/SD conditions, which might offer a solution to the efficacy problem of MSCs transplantation in ischemic heart diseases.

Adiponectin receptor 1 and receptor 2 (AdipoR1 and AdipoR2) have been proved to mainly mediate the action of APN on target tissues [28]. AdipoR1 is abundantly expressed and has high affinity for gAPN, whereas AdipoR2 is predominantly expressed in the liver and has high affinity for fAPN [29]. Binding of APN with AdipoR1 or AdipoR2 leads to the subsequent activation of various signaling pathways, especially the adenosine monophosphate-activated protein kinase (AMPK) pathway [30]. AMPK is a metabolic related protein kinase that regulates energy consumption and production, and activated AMPK limits energy utilization to ensure cell survival [31]. Moreover, AMPK is also crucial in the regulation of MSCs proliferation [32]. In this regard, the present study aimed to investigate the impact of gAPN on the apoptosis of MSCs under H/SD conditions, and further identify the main receptor(s) and the intracellular signaling mechanisms that may mediate such actions, focusing particularly on the AMPK pathways.

Ethics statement

This study was performed in strict accordance with the Chinese guidelines for the care and use of laboratory animals. All animals received humane care and the experimental protocol was approved by the Care of Experimental Animals Committee of Fuwai Hospital.

Cell isolation and culture

Isolation and culture of adult rat bone marrow MSCs were performed as previously described [10]. In brief, bone marrow was harvested from the tibia and femur of Sprague-Dawley rats (60 - 80g, male) and plated into cell culture flasks with complete medium in an incubator set at 37°C containing 5% CO2 and 95% air. The complete medium was consisted of Iscove's Modified Dulbecco's Medium (IMDM, Gibco, USA), 10% fetal bovine serum (Gibco, USA) and 1% penicillin-streptomycin (Gibco, USA). The d-glucose (dextrose) concentration of IMDM is 25 mM. After 24 hours, non-adherent cells were removed by washing with phosphate-buffered saline (PBS). The medium was changed every two to three days thereafter. When the cells reached 80% confluence, they were detached using 0.25% trypsin-EDTA (Gibco, USA) and subcultured at the ratio of 1:2. All cells used in the experiment were passage 3.

Identification of MSCs

Immunophenotyping analysis was employed to identify the surface markers of MSCs. The cells were harvested, washed with PBS, and incubated with fluorescein isothiocyanate (FΙTC)-labeled anti-CD45, phycoerythrin (PE)-conjugated anti-CD29 and Peridinin-Chlorophyll-Protein (PerCP) Cyanine5.5-labeled anti-CD90 (all from eBioscience, USA), as well as allophycocyanin (APC)-labeled anti-CD11b (from Miltenyi Biotec, Germany) antibodies at 4°C for 10 minutes. After washing with PBS, labeled cells were resuspended in 200 µL PBS and analysed with FACS Calibur System (Becton-Dickinson).

Cell treatment

The MSCs were exposed to H/SD conditions for 6 hours as previously described [10]. Briefly, MSCs were washed with PBS twice, and different concentrations (0.01 µg/mL, 0.1 µg/mL, 1 µg/mL) of gAPN (Biovision, USA) were added into the serum free medium. Then MSCs were put into a sealed GENbox hypoxic chamber fitted with an AnaeroPack (Mitsubishi Gas Chemical Company, Japan) to scavenge the free oxygen at 37°C for 6 hours. The oxygen concentration was maintained below 0.1% after 0.5 -1 hour as indicated by an Anaer indicator (Bio-Me'rieux, Marcy I'Etoile, France). MSCs cultured in complete medium under non-hypoxic condition were regarded as the non-apoptotic control group. For pathway studies, the cells were pre-incubated with AMPK inhibitor Compound C (10 µM, Abcam, UK) for 20 minutes before the addition of 1 µg/mL gAPN, and were then exposed to H/SD conditions.

Small interfering RNA-mediated knockdown of AdipoR1 orAdipoR2

Cells were grown in complete medium in a 6-well plate. After reaching approximately 50% confluence, MSCs were transfected with small interfering RNA (siRNA, Genepharma, China) designed to knockdown rat AdipoR1 or AdipoR2 or the combined of them with lipofectmin2000. Scrambled siRNA (Genepharma, China) was transfected as control. The sequences of siRNAs ultimately used for providing optimal efficiency were AdipoR1, sense 5'-GGC UCU AUU ACU CCU UCU ATT-3' and antisense 5'-UAG AAG GAG UAA UAG AGC CTT-3'; AdipoR2, sense 5'-GAG CCA GAU AUA AGG CUC ATT-3' and antisense 5'-UGA GCC UUA UAU CUG GCU CTT-3', and scrambled control, sense 5'-UUC UCC GAA CGU GUC ACG UTT-3' and antisense 5'-ACG UGA CAC GUU CGG AGA ATT-3'.

Assessment of morphological changes

Cell nuclear condensation and fragmentation were assessed using chromatin dye Hoechst 33342 as previously described [10]. Briefly, cells were fixed in 4% paraformaldehyde at room temperature for 30 minutes, and washed with PBS twice, then incubated with 5 mg/L Hoechst 33342 fluorochrome at room temperature (protected from light) for 30 minutes. After rewashing with PBS twice, observations were conducted under a fluorescent microscope (Leica, Germany). Apoptotic cells were identified as fragmented and condensed apoptotic nuclei.

Flow cytometric analysis of cell apoptosis

Cell apoptosis was assessed with flow cytometry using Annexin V-FITC Apoptosis Detection Kit (Becton-Dickinson, USA) in accordance with the manufacturer's instructions. Briefly, after the desired treatments, MSCs were trypsinized and washed once with PBS. Cells were then resuspended in 100 µL 1× binding buffer. Then, 5 µL Annexin V and 5 µL propidium iodide (PI) solution were added to the binding buffer and incubated in the dark at room temperature for 20 minutes. After the staining, 400 µL 1× binding buffer were added to the cell suspension and 1 × 104 stained cells were analyzed using FACS Calibur System (Becton-Dickinson).

Caspase 3 activity

The activity of caspase 3 was measured using the Caspase 3/CPP32 Colorimetric assay kit (Biovision, USA). In brief, after the desired treatment, cells were collected and resuspended in 50 µL of chilled lysis buffer on ice for 10 minutes, and centrifuged at 10,000 g for 1 minute. The supernatant was transferred to a microtube, and the protein concentration was measured using BCA protein assay. Then, 100 µg protein was diluted to 50 µL cell lysis buffer for each assay. Next, 50 µL of 2× Reaction Buffer containing 10 mM DTT were added, followed by adding 5 µL of 4 mM DEVD-pNA substrate (200 µM final concentration) to each sample. After incubation at 37°C for 2 hours, 100 µL of each sample was transferred to a 96-well plate, and the cleavage of the DEVD-pNA was determined by reading the absorbance at 405 nm in a microplate reader.

Measurement of mitochondrial membrane potential (MMP)

The loss of mitochondrial membrane potential was assessed using the fluorescent dye 5, 5', 6, 6'-tetrachloro-1, 1', 3, 3'-tetraethyl benzimidazolylcarbcyanine iodide (JC-1) (Beyotime, China) based on the manufacturer's instructions. In brief, after treatment with indicated agents and exposure to H/SD, MSCs were incubated with 0.5 mL complete medium and 0.5 mL JC-1 staining solution at 37°C for 20 minutes, and then rinsed twice with ice-cold JC-1 staining buffer. The cells were viewed and photographed under a fluorescent microscope (Leica, Germany). To quantify the changes of MMP, the red/green fluorescence intensity was assessed with a fluorescence plate reader. Briefly, MSCs were harvested and resuspended in 0.5 mL complete medium. After incubating with 0.5 mL JC-1 staining solution at 37°C for 20 minutes, MSCs were centrifuged at 600 g for 3 minutes and washed twice with ice-cold JC-1 staining buffer. Then, MSCs were resuspended in 200 µL staining buffer and transferred to a black 96-well plate. Finally, the MMP was determined by the ratio of red over green fluorescent emission in a microplate reader. The excitation and emission wavelengths were 490 nm and 530 nm for detection of monomeric form of JC-1 (green fluorescence), and 525 nm and 590 nm for detection of aggregated form of JC-1 (red fluorescence).

Protein extraction and Western blot analysis

Cells were collected and rinsed with ice-cold PBS twice and then lysed in ice-cold lysis buffer on ice for 30 minutes. Cell lysates were centrifuged at 13,000g at 4°C for 15 minutes and the protein concentrations were determined by the BCA protein assays. Equal amounts of proteins (20 µg/lane) were separated on NuPage 4% -12% Bis-Tris Gels (Novex, Life technologies, USA) by electrophoresis for Western blot analysis. The proteins were then transferred to nitrocellulose membranes using dry electroblotting apparatus (Invitrogen, USA), and the membranes were blocked in 5% skim milk at room temperature for 2 hours. Next, the membranes were incubated with primary antibodies in skim milk over night at 4°C. The primary antibodies used were as follows: β-actin (1:1000), pro-caspase-3 (1:1000), Cl-caspase-3 (1:250), Bcl-2 (1:1000), Bax(1:500), phosphorylated (Thr172)-AMPK (1:500) and AMPK (1:1000). All the above antibodies were from Cell Signal Technology. AdipoR1 (1:500) and AdipoR2 (1:500) antibodies were from Santa Cruz Biotechnology. In the following day the membranes were washed three times and secondary antibodies were added and incubated for 2 hours. After washing, the membranes were processed for analysis using an enhanced chemiluminescence detection system (FluorChem M, USA). The target signals were normalized to the β-actin signal and analyzed semi-quantitatively with Quantity One system.

Statistical analysis

Data are expressed as mean ± standard deviation (SD) from at least 3 independent experiments. Differences among groups were tested by one-way analysis of variance (ANOVA) and Tukey's multiple comparisons via SPSS Statistics software version 22.0. P value less than 0.05 was considered as statistical significance.

Characteristics of MSCs

The MSCs obtained from the bone marrow of the SD rats exhibited a spindle-like appearance. The results of FACS analysis revealed that the majority of adherent cells from passage 3 were positive for CD29 (99.00 ± 0.20%) and CD90 (98.47 ± 0.81%), but were negative for CD45 (1.5 ± 0.53%) and CD11b (1.62 ± 0.11%) (Fig. 1).

Fig. 1

Characteristics of mesenchymal stem cells (MSCs). Cell surface markers were analyzed using flow cytometry. Green line: isotype control; Red line: MSCs staining with CD45-FITC, CD11b-APC, CD90-PerCP Cyanine5.5 and CD29-PE.

Fig. 1

Characteristics of mesenchymal stem cells (MSCs). Cell surface markers were analyzed using flow cytometry. Green line: isotype control; Red line: MSCs staining with CD45-FITC, CD11b-APC, CD90-PerCP Cyanine5.5 and CD29-PE.

Close modal

gAPN dose-dependently protected MSCs from H/SD-induced apoptosis

To delineate the effect of gAPN on the apoptosis of MSCs under H/SD conditions, different concentrations of gAPN were added into the serum-free medium, and apoptosis was determined morphologically. As shown in Fig. 2A, cells exposed to H/SD conditions appeared to have shrunken and condensed nuclei. In contrast, inclusion of gAPN dose-dependently reduced the nuclear change induced by H/SD, with 1 µg/mL closely reaching the control level.

Fig. 2

Effects of globular adiponectin (gAPN) on hypoxia/serum deprivation (H/SD)-induced apoptosis in MSCs. (A) Nuclear morphology was determined using Hoechst 33342 staining. Representative photo-micrographs showed the apoptotic nuclear condensation (white arrows) in MSCs (100×). (B-C) Apoptosis was quantified by FACS analysis after staining with Annexin V and propidine iodine (PI). The Annexin V+/ PI- cells indicated the early apoptotic cells, while the Annexin V+/ PI+ cells were the late apoptotic cells. (D) Quantitative analysis of caspase-3 activity was conducted using CPP32 Colorimetric assay. Changes in activation of caspase-3 were expressed as fold increase relative to the control values. Each column represents mean ± SD of three independent experiments. *p < 0.05 versus control group; #p < 0.05 versus H/SD group.

Fig. 2

Effects of globular adiponectin (gAPN) on hypoxia/serum deprivation (H/SD)-induced apoptosis in MSCs. (A) Nuclear morphology was determined using Hoechst 33342 staining. Representative photo-micrographs showed the apoptotic nuclear condensation (white arrows) in MSCs (100×). (B-C) Apoptosis was quantified by FACS analysis after staining with Annexin V and propidine iodine (PI). The Annexin V+/ PI- cells indicated the early apoptotic cells, while the Annexin V+/ PI+ cells were the late apoptotic cells. (D) Quantitative analysis of caspase-3 activity was conducted using CPP32 Colorimetric assay. Changes in activation of caspase-3 were expressed as fold increase relative to the control values. Each column represents mean ± SD of three independent experiments. *p < 0.05 versus control group; #p < 0.05 versus H/SD group.

Close modal

Apoptotic cells were then quantified using flow cytometry after Annexin V and PI staining. The Annexin V-FITC assay detected the exposure of phosphatidylserine (PS) to the external surface of the plasma membrane in early apoptosis, whereas the PI assay primarily measured late apoptotic and necrotic cells [33]. The early apoptotic cells (Annexin V+ ⁄ PI-), late apoptotic cells (Annexin V+ ⁄ PI+) and necrotic cells (Annexin V- ⁄ PI+) cells were thus discriminated. Flow cytometry results indicated that, compared with the control group, H/SD significantly increased the percentage of both early (9.05 ± 1.20% vs. 1.13 ± 0.42% in control group, p < 0.05) and late (13.03 ± 0.91% vs. 3.42 ± 0.65% in control group, p < 0.05) apoptotic MSCs. Conversely, gAPN treatment concentration-dependently led to a decrease in apoptotic cells induced by H/SD. The anti-apoptotic effect reached its peak at 1 µg/mL gAPN, with a reduction in early (1.56 ± 0.27% vs. 9.05 ± 1.20% in H/SD group, p < 0.05) and late (4.54 ± 0.96% vs. 13.03 ± 0.91% in H/SD group, p < 0.05) apoptotic cells (Fig. 2B, C).

A typical feature of apoptosis is the activation of caspases, a family of cysteine proteases that plays an essential role in this process. To specify the involvement of gAPN in the survival of MSCs after H/SD, the changes of caspase-3 activity with reference to the control values were analyzed. Results showed that exposure of MSCs to H/SD brought about an approximately 4 to 5 fold increase in the caspase-3 activity, which was inhibited by gAPN in a dose-dependent manner, with 1 µg/mL almost reaching that of the control group (Fig. 2D). The activation of caspase-3 was further expressed as the cleavage of pro-caspase-3 into its active subunit, cleaved caspase-3 (Cl-caspase-3), using the Western blot analysis (Fig. 3A). Compared to the H/SD group, the cleavage of pro-caspase-3 was markedly decreased with the addition of gAPN (Fig. 3B, C). These data suggested that gAPN dose-dependently protected MSCs from apoptosis induced by H/SD, and the maximum effect was observed at 1 µg/mL gAPN, which was used in subsequent experiments.

Fig. 3

Effects of gAPN on the transition of pro-caspase-3 to cleaved -caspase-3. (A) The expressions of procaspase-3 and active subunit of cleaved-caspase-3 were detected by Western blot. (B-C) Quantitative analysis of the expressions of pro-caspase-3 and cleaved-cas-pase-3 relative to β-actin was shown in the bar graphs. Each column represents mean ± SD of three independent experiments. * p < 0.05 versus control group; # p < 0.05 versus H/SD group.

Fig. 3

Effects of gAPN on the transition of pro-caspase-3 to cleaved -caspase-3. (A) The expressions of procaspase-3 and active subunit of cleaved-caspase-3 were detected by Western blot. (B-C) Quantitative analysis of the expressions of pro-caspase-3 and cleaved-cas-pase-3 relative to β-actin was shown in the bar graphs. Each column represents mean ± SD of three independent experiments. * p < 0.05 versus control group; # p < 0.05 versus H/SD group.

Close modal

AdipoR1 mainly mediated the anti-apoptotic effects of gAPN

To investigate the main type of receptor that mediated the actions of gAPN, siRNAs targeting AdipoR1 or AdipoR2 or both of them were used. The knockdown efficiency was confirmed by evaluating the expressions of these proteins using Western blot. As shown in Fig. 4, AdipoR1 and AdipoR2 expressions were significantly suppressed after using corresponding siRNA but not with scrambled siRNA or the unspecific siRNA. Then flow cytometry and caspase-3 activity kit were employed as quantitative assays to decide the functional significance of manipulating AdipoR1 or AdipoR2 expression. As displayed in Fig. 5A-B, compared with the scrambled group, the knockdown of AdipoR1 resulted in a significant increase in apoptotic cells at early (8.12 ± 1.64% vs. 1.58 ± 0.21% in scrambled group, p < 0.05) and late (8.11 ± 1.98% vs. 4.61 ± 0.85% in scrambled group, p < 0.05) phases, whereas no obvious changes were observed with the knockdown of AdipoR2 at early (2.25 ± 0.31% vs. 1.58 ± 0.21% in scrambled group, p > 0.05) and late (4.85 ± 0.97% vs. 4.61 ± 0.85% in scrambled group, p > 0.05) phases. Suppression of both AdipoR1 and AdipoR2 expression in combination by siRNA almost abolished the protective effect of gAPN against early (8.73 ± 1.47% vs. 1.58 ± 0.21% in scrambled group, p < 0.05; 8.73 ± 1.47% vs. 9.05 ± 1.20% in H/SD group, p > 0.05) and late (11.33 ± 0.91% vs. 4.61 ± 0.85% in scrambled group, p < 0.05; 11.33 ± 0.91% vs. 13.03 ± 0.91% in H/SD group, p > 0.05) apoptosis. The differences between si-AdipoR1 group and combined knockdown of AdipoR1 and AdipoR2 group were not statistically significant. In parallel, the caspase-3 activity had a greater increase with the knockdown of AdipoR1 (2.91 ± 0.64 vs. 1.08 ± 0.08 in scrambled group, p < 0.05) than that of AdipoR2 (1.44 ± 0.32 vs. 1.08 ± 0.08 in scrambled group, p > 0.05). The combined knockdown of AdipoR1 and AdipoR2 almost reversed the suppressive effect of gAPN on caspase-3 activity (3.84 ± 0.18 vs. 1.08 ± 0.08 in scrambled group, p < 0.05; 3.84 ± 0.18 vs. 4.65 ± 0.84 in H/SD group, p > 0.05) (Fig. 5C). These findings indicate that it was AdipoR1, rather than AdipoR2 that mainly mediated the anti-apoptosis effect of gAPN against H/SD conditions.

Fig. 4

The knockdown efficiency of adiponectin receptor 1 (AdipoR1) and receptor 2 (AdipoR2) were determined by Western blot. Each column represents mean ± SD of three independent experiments. *p < 0.05 versus scrambled siRNA group.

Fig. 4

The knockdown efficiency of adiponectin receptor 1 (AdipoR1) and receptor 2 (AdipoR2) were determined by Western blot. Each column represents mean ± SD of three independent experiments. *p < 0.05 versus scrambled siRNA group.

Close modal
Fig. 5

Roles of AdipoR1 and AdipoR2 in gAPN-mediated anti-apoptotic effect under H/SD conditions. (A-B) Apoptosis was quantified by FACS analysis of Annexin V and PI positive cells after the knockdown of AdipoR1 or AdipoR2 or both of them siRNA. (C) Chances in caspase-3 activity after the knockdown AdipoR1 or AdipoR2 both of them, determined by colorimetric assay, were shown as fold increase relative to the control values. Each column represents mean ± SD of three independent experiments. * P < 0.05 versus control group; # P < 0.05 versus H/SD group; $ P < 0.05 versus H/SD+gAPN+scrambled siRNA group.

Fig. 5

Roles of AdipoR1 and AdipoR2 in gAPN-mediated anti-apoptotic effect under H/SD conditions. (A-B) Apoptosis was quantified by FACS analysis of Annexin V and PI positive cells after the knockdown of AdipoR1 or AdipoR2 or both of them siRNA. (C) Chances in caspase-3 activity after the knockdown AdipoR1 or AdipoR2 both of them, determined by colorimetric assay, were shown as fold increase relative to the control values. Each column represents mean ± SD of three independent experiments. * P < 0.05 versus control group; # P < 0.05 versus H/SD group; $ P < 0.05 versus H/SD+gAPN+scrambled siRNA group.

Close modal

AMPK was a pivotal mediator of gAPN suppression of apoptosis

APN exerts its action in various cell systems by activating AMPK signaling pathways [14,18,30,34,35,36]. To determine whether this signaling pathway was stimulated by gAPN in MSCs, phosphorylation of AMPK was assessed following treatment with different concentrations of gAPN under H/SD conditions for 6 hours. Results showed that gAPN induced a dose-dependent increase of phosphorylation of AMPK (p-AMPK) at Thr-172 with 1 µg/mL gAPN reached its peak, and this effect was significantly inhibited by pretreatment with Compound C (10 µM) (0.51 ± 0.09 vs. 1.25 ± 0.15 in 1 µg/mL gAPN group, p < 0.05) (Fig. 6A). Further, compared with the scrambled group, the knockdown of AdipoR1 partly inhibited the p-AMPK (0.75 ± 0.13 vs. 1.25 ± 0.15 in scrambled group, p < 0.05) (Fig. 6B).

Fig. 6

Effects of gAPN on phosphorylation of adenosine monophosphate-activated protein kinase (AMPK) in MSCs. (A) Representative Western blots of phosphorylated and total AMPK in MSCs that were treated with different concentrations of gAPN alone or pretreated with Compound C (10 µM) for 20 minutes. (B) Representative Western blots of phosphorylated AMPK in MSCs that were transfected with scrambled, AdipoR1 or AdipoR2 siRNAs before exposure to H/SD conditions for 6 hours. Each column represents mean ± SD of three independent experiments. * p < 0.05 versus control group; # p < 0.05 versus H/SD group; $ p < 0.05 versus H/SD + gAPN (+ scrambled siRNA) group.

Fig. 6

Effects of gAPN on phosphorylation of adenosine monophosphate-activated protein kinase (AMPK) in MSCs. (A) Representative Western blots of phosphorylated and total AMPK in MSCs that were treated with different concentrations of gAPN alone or pretreated with Compound C (10 µM) for 20 minutes. (B) Representative Western blots of phosphorylated AMPK in MSCs that were transfected with scrambled, AdipoR1 or AdipoR2 siRNAs before exposure to H/SD conditions for 6 hours. Each column represents mean ± SD of three independent experiments. * p < 0.05 versus control group; # p < 0.05 versus H/SD group; $ p < 0.05 versus H/SD + gAPN (+ scrambled siRNA) group.

Close modal

We extended our study by examining whether activation of this pathway was critical for the anti-apoptotic actions of gAPN in MSCs. Results showed that pre-incubation with Compound C partly suppressed the anti-apoptotic effect of gAPN on MSCs, evidenced by increased early apoptotic cells (6.38 ± 0.59% vs. 1.55 ± 0.27% in gAPN group, p < 0.05) and late apoptotic cells (8.83 ± 1.96% vs. 4.54 ± 0.96% in gAPN group, p < 0.05) (Fig. 7A, B), as well as elevated caspase-3 activity (3.22 ± 0.29 vs. 1.07 ± 0.2 in gAPN group, p < 0.05) (Fig. 7C). These results illustrated that gAPN inhibited the apoptosis of MSCs against H/SD conditions at least partly through the AMPK pathway.

Fig. 7

Role of AMPK in gAPN-mediated anti-apoptotic effect on MSCs under H/SD conditions. (A-B) Apoptosis was quantified by FACS analysis of Annexin V and PI positive cells after the inhibition of AMPK pathway using Compound C. (C) Changes in caspase-3 activity after inhibiting the phosphorylation of AMPK, determined by colorimetric assay, were shown as fold increase relative to the control values. Each column represents mean ± SD of three independent experiments. * p < 0.05 versus control group; # p < 0.05 versus H/SD group; $ p < 0.05 versus H/SD + gAPN group.

Fig. 7

Role of AMPK in gAPN-mediated anti-apoptotic effect on MSCs under H/SD conditions. (A-B) Apoptosis was quantified by FACS analysis of Annexin V and PI positive cells after the inhibition of AMPK pathway using Compound C. (C) Changes in caspase-3 activity after inhibiting the phosphorylation of AMPK, determined by colorimetric assay, were shown as fold increase relative to the control values. Each column represents mean ± SD of three independent experiments. * p < 0.05 versus control group; # p < 0.05 versus H/SD group; $ p < 0.05 versus H/SD + gAPN group.

Close modal

gAPN elicited anti-apoptotic effect via the inhibition of the mitochondrial-related intrinsic pathway

The balance of anti-apoptotic protein Bcl-2 and pro-apoptotic protein Bax plays an important role in the regulation of mitochondrial integrity and cell survival [37]. To ascertain whether the mitochondrial-dependent apoptotic pathway was involved in the gAPN's protective effect on MSCs, the effect of gAPN on H/SD-induced Bcl-2 and Bax expression changes were examined. Western blot analysis revealed that H/SD significantly decreased Bcl-2 (0.51 ± 0.20 vs. 1.50 ± 0.25 in control group, p < 0.05) and increased Bax (1.29 ± 0.06 vs. 0.80 ± 0.26 in control group, p < 0.05) expressions. Compared to the H/SD group, 1 µg/mL gAPN-treated group significantly increased expression of Bcl-2 (1.42 ± 0.32 vs. 0.51 ± 0.20 in H/SD group, p < 0.05) and decreased expression of Bax (0.70 ± 0.21 vs. 1.29 ± 0.06 in H/SD group, p < 0.05) (Fig. 8A). In addition, the gAPN-induced up-regulation of Bcl-2 and down-regulation of Bax expressions were markedly attenuated with the knockdown of AdipoR1 and inhibition of p-AMPK by Compound C (Fig. 8B, C).

Fig. 8

Effects of gAPN on the expressions of Bcl-2 and Bax relative to β-actin were detected by Western blot. (A) The expressions of Bcl-2 and Bax in MSCs treated with different concentrations of gAPN. (B) The expressions of Bcl-2 and Bax after the knockdown of AdipoR1 or AdipoR2 using siRNA. (C) The expressions of Bcl-2 and Bax after inhibiting the phosphorylation of AMPK with Compound C. Each column represents mean ± SD of three independent experiments. * p < 0.05 versus control group; # p< 0.05 versus H/SD group; $ p < 0.05 versus H/SD + gAPN (+ scrambled siRNA) group.

Fig. 8

Effects of gAPN on the expressions of Bcl-2 and Bax relative to β-actin were detected by Western blot. (A) The expressions of Bcl-2 and Bax in MSCs treated with different concentrations of gAPN. (B) The expressions of Bcl-2 and Bax after the knockdown of AdipoR1 or AdipoR2 using siRNA. (C) The expressions of Bcl-2 and Bax after inhibiting the phosphorylation of AMPK with Compound C. Each column represents mean ± SD of three independent experiments. * p < 0.05 versus control group; # p< 0.05 versus H/SD group; $ p < 0.05 versus H/SD + gAPN (+ scrambled siRNA) group.

Close modal

As the loss of mitochondrial membrane potential (MMP) is regarded as one of the inducers of mitochondrial-related apoptosis, whether gAPN influenced the loss of MMP in MSCs subjected to H/SD conditions was examined using JC-1 fluorescence staining assay. JC-1 exists as an aggregated form (red fluorescence) in the matrix of mitochondria with the normal MMP, and it is converted to the monomeric form (green fluorescence) with the loss of MMP. Consistent with the anti-apoptotic effect, treatment with 1 µg/mL gAPN led to a strong protection from MMP loss induced by H/SD, as determined by the reduction of green fluorescence and increase of red fluorescence in gAPN group compared to the H/SD group (Fig. 9A). To quantify the changes of MMP, the red/green fluorescence intensity was assessed with a fluorescence plate reader. Compared with the control group, H/SD induced a significant reduction of red/green fluorescence (0.68 ± 0.05 fold vs. control group, p < 0.05). The addition of gAPN significantly reversed the above change, with the red/green fluorescence closely reaching the control level (0.97 ± 0.04 vs. 0.68 ± 0.05 in H/SD group, p < 0.05). Moreover, the inhibition of the AdipoR1 (0.72 ± 0.04 vs. 0.93 ± 0.05 in scrambed group, p < 0.05) or AMPK pathway (0.82 ± 0.03 vs. 0.97 ± 0.04 in gAPN group, p < 0.05) partly suppressed the protective effect of gAPN on MMP expressed as the red/green fluorescence (Fig. 9B, C).

Fig. 9

Effects of gAPN on the changes of mitochondrial membrane potential (MMP) detected with JC-1 fluorescence staining assay. The red fluorescence represents JC-1 aggregates in the matrix of mitochondria with the normal MMP, while the green fluorescence represents JC-1 monomer appeared with the depolarization of mitochondrial membrane. Loss of MMP was expressed as conversion of red fluorescence to green fluorescence. (A) Representative images of merged fluorescence images from each group photographed by the fluorescent microscope (100×). (B-C) Quantitative analyses of the red/green fluorescence ratio were conducted with a fluorescence microplate reader. Red/green fluorescence ratios were expressed as fold changes relative to the control group. Each column represents mean ± SD of three independent experiments. * p < 0.05 versus control group; # p < 0.05 versus H/SD group; $ p < 0.05 versus H/SD + gAPN (+ scrambled siRNA) group.

Fig. 9

Effects of gAPN on the changes of mitochondrial membrane potential (MMP) detected with JC-1 fluorescence staining assay. The red fluorescence represents JC-1 aggregates in the matrix of mitochondria with the normal MMP, while the green fluorescence represents JC-1 monomer appeared with the depolarization of mitochondrial membrane. Loss of MMP was expressed as conversion of red fluorescence to green fluorescence. (A) Representative images of merged fluorescence images from each group photographed by the fluorescent microscope (100×). (B-C) Quantitative analyses of the red/green fluorescence ratio were conducted with a fluorescence microplate reader. Red/green fluorescence ratios were expressed as fold changes relative to the control group. Each column represents mean ± SD of three independent experiments. * p < 0.05 versus control group; # p < 0.05 versus H/SD group; $ p < 0.05 versus H/SD + gAPN (+ scrambled siRNA) group.

Close modal

The therapeutic potential of MSCs for cardiac repair is hampered partly due to their low survival rate within the ischemic myocardial microenvironment into which MSCs are introduced [8]. Hypoxia and serum deprivation, imitating the ischemic engraftment environment, induced the apoptosis of MSCs through the mitochondrial apoptotic pathway [10]. This study found that gAPN could confer a protective effect against the above-mentioned process after binding with AdipoR1 and activating the downstream AMPK pathway. These results highlight an opportunity to improve the survival and therapeutic efficacy of MSCs by targeting gAPN.

APN is an adipocyte-secreted protein that elicits protective effects in the vasculature and myocardium with anti-diabetic, anti-atherosclerotic and anti-apoptotic properties [17]. It is proposed as a tissue-regenerating hormone and regulates the survival, migration and differentiation of several types of stem cells [21]. gAPN, the globular C-terminal domain of APN, is biologically active and easier to manufacture and administer [20,23,24,38]. Our results indicate that gAPN may enhance the survival of MSCs under H/SD conditions, potentially benefit the MSCs transplantation efficacy in addition to the cardioprotective effects. There is indeed precedence suggesting that APN serves as a prosurvival stem cell factor. It was reported that APN promoted the survival of bone marrow mononuclear cells (BM-MNCs) against apoptosis triggered by staurosporine or ceramide [22]. gAPN also counteracted the apoptotic process triggered by growth factor withdrawal in murine mesoangioblasts [24]. Besides, gAPN increased the endothelial repair and angiogenesis by increasing the number and function of endothelial progenitor cells (EPCs) [39,40]. In addition, Hou M et al. showed that C1q tumor necrosis factor-related protein-3 (CTRP3), a newly identified adipokine similar to APN, protected MSCs against H/SD-induced apoptosis through the PI3K/Akt pathway [41]. However, the up-stream molecular / receptor that mediates the action between CTRP3 and PI3K has not been investigated.

In the present study, hypoxia and serum deprivation were combined to imitate the ischemic environment and serve as the inducer of apoptosis in MSCs. It was found that serum deprivation was the predominant factor in this process [10], while hypoxia alone failed to induce apoptosis [32] and exposure to 1-1.5% O2 even enhanced the proliferation of MSCs [42,43]. The resistance to hypoxia per se is largely due to the up-regulated glycolytic activities as indicated by the high-level expression of glycolytic enzymes in MSCs [44]. Effects of hypoxia are usually mediated by hypoxia inducible factor-1 (HIF-1). It was verified that over-expression of HIF-1alpha conferred resistance to hypoxia and oxidative stress-induced apoptosis in bone marrow-derived MSCs [45]. Wagegg M et al. found that hypoxia (less than 2% O2) resulted in a significant time-dependent increase in the expressions of HIF-1alpha mRNA and protein in bone marrow-derived MSCs after 72 hours and 2 weeks of incubation when compared to cells under normoxia. However, there were no detectable protein expression of HIF-1 in both normoxia and hypoxia conditions within 24 hours [46]. As the exposure time of MSCs to hypoxia condition was 6 hours in our study, not long enough to induce the expression of HIF-1, and the main contributer of apoptosis is serum deprivation instead of hypoxia [10], HIF-1 might have little involvement in the anti-apoptotic effect of gAPN in our settings. Under serum deprivation condition, APN also exerts anti-apoptotic effects on endothelial cells [34] and ventricular myocytes [14], which may facilitate cardiac repair to a larger extent. Nevertheless, there were studies pointing out that APN could induce apoptosis in some cancer cell lines such as myelomonocytic leukemia cells [47], breast cancer cells [48] and endometrial cancer cells [49]. These findings suggested that the effects of APN on cell survival are highly dependent on cellular type [50].

It was observed in this study that targeted knockdown of AdipoR1 by specific siRNA significantly inhibited the protective effect of gAPN on H/SD-induced apoptosis of MSCs, while AdipoR2 siRNA only slightly suppressed the anti-apoptotic effect. The combined knockdown of AdipoR1 and AdipoR2 almost eliminated the protective effect of gAPN, although this effect was not statistically significant with the only knockdown of AdipoR1. Further, previous studies have shown that gAPN has higher affinity for AdipoR1 receptor subtype [28,51]. On top of that, the mRNA expression level of AdipoR1 was greater than that of AdipoR2 in MSCs, and AdipoR1 played a key role in mediating APN signaling cascade for the regulation of osteoblast differentiation in MSCs [52]. In this regard, it is reasonable to conclude that AdipoR1, rather than AdipoR2, was the main type of receptor mediating the anti-apoptotic effect of gAPN in our study. Our results correlate well with the previous reports suggesting that AdipoR1 primarily mediated the protective effects of gAPN on the H9C2 cells from hypoxia/reoxygenation-induced apoptosis [20]. It was also demonstrated that the metabolic effects of gAPN in primary neonatal cardiomyocytes were mediated via AdipoR1 [53]. In addition, AdipoR1 also mediated the gAPN's unregulation of EPCs function suppressed by high glucose [39].

Binding of gAPN with its specific receptor generates various signaling pathways that regulate cell survival and apoptosis, especially the AMPK pathway. The activation of AMPK by gAPN is critical in cellular responses to metabolic stress involving energy generation and consumption [30,36,54,55]. Under hypoxia, AMPK is normally activated and functions to maintain the balance of catabolism and anabolism in cells like cardiomyocytes [56], which re-establishes energy balance and enhances cell survival in the absence of oxygen. By contrast, hypoxic condition did not alter the phosphorylation of AMPK in MSCs, which was correlated with the maintenance of ATP concentration [32]. In line with the previous results, our data also revealed that hypoxia and serum deprivation alone did not activate AMPK, while gAPN induced sustained phosphorylation of AMPK in MSCs. Further, the application of Compound C, the inhibitor of AMPK, significantly inhibited the protection offered by gAPN against H/SD-induced apoptosis. Thus, taken together, these findings indicate that AMPK pathway might be involved in the anti-apoptotic actions of gAPN. However, Compound C was reported to have some AMPK-independent actions even though it was widely used to verify the role of AMPK in various (patho)-physiological processes [57,58]. Hence, the knockdown of AMPKalpha1 (the sole catalytic isoform expressed in MSCs) by RNA-silencing might provide more potent evidence of the role AMPK played in the above-mentioned process.

Our study is consistent with the previous reports showing that AMPK mediated the protective effects of APN on the apoptosis of endothelial cells induced by serum starvation [34]. Moreover, data suggested that APN counteracted ventricular myocytes and fibroblasts apoptosis triggered by serum deprivation and hypoxia-reoxygenation through AMPK-dependent mechanism [14]. However, there are reports pointing out that the gAPN significantly reduced cell death of cardiomyocytes induced by simulated ischemia and reperfusion in an AMPK-independent fashion [59]. It therefore remains to be elucidated whether the AMPK-dependent anti-apoptotic action of APN is dependent on cell types or different forms of APN.

It has been established that H/SD induces MSCs apoptosis through the mitochondrial-related intrinsic pathway, leading to the accumulation of Bax, loss in mitochondrial membrane potential (MMP) and activation of caspase-3 [10]. The balance of Bax and Bcl-2 expressions play a key role in maintaining the integrity of mitochondrial and suppressing the mitochondrial apoptotic pathway [60,61]. The decrease of Bcl-2 [62] and increase of Bax [63,64] lead to loss of MMP and release of cytochrome c from the mitochondria to the cytoplasm, which activates the caspase-3 effector and leads to apoptotic cell death [37,65]. Importantly, APN was found to reverse neuroblastoma cell apoptosis via regulation of Bcl-2 and Bax expression [61,66] and attenuate the mitochondrial dysfunction in diabetic heart [54].

Mitochondria in undifferentiated MSCs were maintained at a relatively low activity status [44]. Nevertheless, it was found to be highly sensitive to external environment such as changes in the energy status, oxidative stress and apoptotic stimuli [67]. Under H/SD conditions, MSCs exhibited mitochondrial dysfunction with MMP loss [10]. This was confirmed by other studies showing that the mitochondrial-dependent pathway exerted a vital function in other molecules and medications protecting MSCs from apoptosis induced by H/SD, including CTRP3 [41], lysophosphatidic acid [68], angiopoietin-1 [69], lovastatin [70], Tongxinluo [71] and Exendin-4 [72].

Our data showed that Bcl-2 was up-regulated and Bax was down-regulated in proportion to the dose of gAPN and AMPK phosphorylation. On the ground that the ratio between Bcl-2 and Bax proteins plays a pivotal role in the apoptotic cascade, it is not surprising that treatment with gAPN was also associated with the prevention of the downstream loss of MMP, finally preventing caspase-3 activation and apoptotic signaling pathways. These results indicated that cytoprotection of gAPN was mediated through the inhibition of the mitochondrial apoptotic pathway. Our results are in accordance with the protective effects of APN on mitochondrial depolarization and apoptosis of mesoangioblasts induced by growth factors withdrawal [24].

Our study reveals that gAPN protects MSCs against the mitochondrial-related apoptosis under H/SD conditions, which is mainly mediated through the AdipoR1 linked to the downstream AMPK signaling pathways. These findings suggest that gAPN may be a novel survival factor for MSCs in the ischemic engraftment environment and further studies in vivo are needed to verify that gAPN might enhance the therapeutic efficacy of MSCs transplantation in ischemic heart diseases.

MSCs (Mesenchymal stem cells); BM-MNCs (Bone marrow mononuclear cells); HSCs (Hemopoietic stem cells); gAPN (globular adiponectin); AMPK (AMP-activated protein kinase); H/SD (Hypoxia and serum deprivation); AdipoR1 (Adiponectin receptor 1); AdipoR2 (Adiponectin receptor 2); MMP (Mitochondrial membrane potential).

This work was supported by grants from the National Natural Science Foundation of China (81170129,81200107,81300157,81200108),863 Program of China (2011AA020110, 2013AA020101), 973 Program of China (2012CB518602) and the Ph.D Programs Foundation of Ministry of Education of China (20111106110012).

The authors have no potential conflict of interests to declare.

1.
Jeevanantham V, Butler M, Saad A, Abdel-Latif A, Zuba-Surma EK, Dawn B: Adult bone marrow cell therapy improves survival and induces long-term improvement in cardiac parameters: a systematic review and meta-analysis. Circulation 2012;126:551-568.
2.
Karantalis V, Hare JM: Use of mesenchymal stem cells for therapy of cardiac disease. Circ Res 2015;116:1413-1430.
3.
Zhang Y, Liang X, Lian Q, Tse HF: Perspective and challenges of mesenchymal stem cells for cardiovascular regeneration. Expert Rev Cardiovasc Ther 2013;11:505-517.
4.
Narita T, Suzuki K: Bone marrow-derived mesenchymal stem cells for the treatment of heart failure. Heart Fail Rev 2015;20:53-68.
5.
Perin EC, Borow KM, Silva GV, DeMaria AN, Marroquin OC, Huang P, Traverse JH, Krum H, Skerrett D, Zheng Y, Willerson JT, Itescu S, Henry TD: A phase II dose-escalation study of allogeneic mesenchymal precursor cells in patients with ischemic or non-ischemic heart failure. Circ Res 2015;117:576-584.
6.
Hare JM, Fishman JE, Gerstenblith G, DiFede Velazquez DL, Zambrano JP, Suncion VY, Tracy M, Ghersin E, Johnston PV, Brinker JA, Breton E, Davis-Sproul J, Schulman IH, Byrnes J, Mendizabal AM, Lowery MH, Rouy D, Altman P, Wong Po Foo C, Ruiz P, Amador A, Da Silva J, McNiece IK, Heldman AW, George R, Lardo A: Comparison of allogeneic vs autologous bone marrow-derived mesenchymal stem cells delivered by transendocardial injection in patients with ischemic cardiomyopathy: the POSEIDON randomized trial. JAMA 2012;308:2369-2379.
7.
Williams AR, Trachtenberg B, Velazquez DL, McNiece I, Altman P, Rouy D, Mendizabal AM, Pattany PM, Lopera GA, Fishman J, Zambrano JP, Heldman AW, Hare JM: Intramyocardial stem cell injection in patients with ischemic cardiomyopathy: functional recovery and reverse remodeling. Circ Res 2011;108:792-796.
8.
Geng YJ: Molecular mechanisms for cardiovascular stem cell apoptosis and growth in the hearts with atherosclerotic coronary disease and ischemic heart failure. Ann N Y Acad Sci 2003;1010:687-697.
9.
Segers VF, Lee RT: Stem-cell therapy for cardiac disease. Nature 2008;451:937-942.
10.
Zhu W, Chen J, Cong X, Hu S, Chen X: Hypoxia and serum deprivation-induced apoptosis in mesenchymal stem cells. Stem Cells 2006;24:416-425.
11.
Fisman EZ, Tenenbaum A: Adiponectin: a manifold therapeutic target for metabolic syndrome, diabetes, and coronary disease? Cardiovasc Diabetol 2014;13:103.
12.
Fruebis J, Tsao TS, Javorschi S, Ebbets-Reed D, Erickson MR, Yen FT, Bihain BE, Lodish HF: Proteolytic cleavage product of 30-kDa adipocyte complement-related protein increases fatty acid oxidation in muscle and causes weight loss in mice. Proc Natl Acad Sci USA 2001;98:2005-2010.
13.
Whitehead JP, Richards AA, Hickman IJ, Macdonald GA, Prins JB: Adiponectin-a key adipokine in the metabolic syndrome. Diabetes Obes Metab 2006;8:264-280.
14.
Shibata R, Sato K, Pimentel DR, Takemura Y, Kihara S, Ohashi K, Funahashi T, Ouchi N, Walsh K: Adiponectin protects against myocardial ischemia-reperfusion injury through AMPK- and COX-2-dependent mechanisms. Nat Med 2005;11:1096-1103.
15.
Shibata R, Izumiya Y, Sato K, Papanicolaou K, Kihara S, Colucci WS, Sam F, Ouchi N, Walsh K: Adiponectin protects against the development of systolic dysfunction following myocardial infarction. J Mol Cell Cardiol 2007;42:1065-1074.
16.
Han SH, Quon MJ, Kim JA, Koh KK: Adiponectin and cardiovascular disease: response to therapeutic interventions. J Am Coll Cardiol 2007;49:531-538.
17.
Goldstein BJ, Scalia RG, Ma XL: Protective vascular and myocardial effects of adiponectin. Nat Clin Pract Cardiovasc Med 2009;6:27-35.
18.
Kim JE, Song SE, Kim YW, Kim JY, Park SC, Park YK, Baek SH, Lee IK, Park SY: Adiponectin inhibits palmitate-induced apoptosis through suppression of reactive oxygen species in endothelial cells: involvement of cAMP/protein kinase A and AMP-activated protein kinase. J Endocrinol 2010;207:35-44.
19.
Lin LY, Lin CY, Su TC, Liau CS: Angiotensin II-induced apoptosis in human endothelial cells is inhibited by adiponectin through restoration of the association between endothelial nitric oxide synthase and heat shock protein 90. FEBS Lett 2004;574:106-110.
20.
Park M, Youn B, Zheng XL, Wu D, Xu A, Sweeney G: Globular adiponectin, acting via AdipoR1/APPL1, protects H9c2 cells from hypoxia/reoxygenation-induced apoptosis. PLoS One 2011;6:e19143.
21.
Fiaschi T, Magherini F, Gamberi T, Modesti PA, Modesti A: Adiponectin as a tissue regenerating hormone: more than a metabolic function. Cell Mol Life Sci 2014;71:1917-1925.
22.
Eren P, Camus S, Matrone G, Ebrahimian TG, Francois D, Tedgui A, Sebastien Silvestre J, Blanc-Brude OP: Adiponectinemia controls pro-angiogenic cell therapy. Stem Cells 2009;27:2712-2721.
23.
Fiaschi T, Giannoni E, Taddei ML, Chiarugi P: Globular adiponectin activates motility and regenerative traits of muscle satellite cells. PLoS One 2012;7:e34782.
24.
Fiaschi T, Tedesco FS, Giannoni E, Diaz-Manera J, Parri M, Cossu G, Chiarugi P: Globular adiponectin as a complete mesoangioblast regulator: role in proliferation, survival, motility, and skeletal muscle differentiation. Mol Biol Cell 2010;21:848-859.
25.
DiMascio L, Voermans C, Uqoezwa M, Duncan A, Lu D, Wu J, Sankar U, Reya T: Identification of adiponectin as a novel hemopoietic stem cell growth factor. J Immunol 2007;178:3511-3520.
26.
Zhang D, Guo M, Zhang W, Lu XY: Adiponectin stimulates proliferation of adult hippocampal neural stem/progenitor cells through activation of p38 mitogen-activated protein kinase (p38MAPK)/glycogen synthase kinase 3beta (GSK-3beta)/beta-catenin signaling cascade. J Biol Chem 2011;286:44913-44920.
27.
Yu L, Tu Q, Han Q, Zhang L, Sui L, Zheng L, Meng S, Tang Y, Xuan D, Zhang J, Murray D, Shen Q, Cheng J, Kim SH, Dong LQ, Valverde P, Cao X, J. C: Adiponectin regulates bone marrow mesenchymal stem cell niche through a unique signal transduction pathway - an approach for treating bone disease in diabetes. Stem Cells 2015;33:240-252.
28.
Yamauchi T, Kamon J, Ito Y, Tsuchida A, Yokomizo T, Kita S, Sugiyama T, Miyagishi M, Hara K, Tsunoda M, Murakami K, Ohteki T, Uchida S, Takekawa S, Waki H, Tsuno NH, Shibata Y, Terauchi Y, Froguel P, Tobe K, Koyasu S, Taira K, Kitamura T, Shimizu T, Nagai R, Kadowaki T: Cloning of adiponectin receptors that mediate antidiabetic metabolic effects. Nature 2003;423:762-769.
29.
Kadowaki T, Yamauchi T, Kubota N, Hara K, Ueki K, Tobe K: Adiponectin and adiponectin receptors in insulin resistance, diabetes, and the metabolic syndrome. J Clin Invest 2006;116:1784-1792.
30.
Konishi M, Haraguchi G, Ohigashi H, Ishihara T, Saito K, Nakano Y, Isobe M: Adiponectin protects against doxorubicin-induced cardiomyopathy by anti-apoptotic effects through AMPK up-regulation. Cardiovasc Res 2011;89:309-319.
31.
McGee SL, Hargreaves M: AMPK and transcriptional regulation. Front Biosci 2008;13:3022-3033.
32.
de Meester C, Timmermans AD, Balteau M, Ginion A, Roelants V, Noppe G, Porporato PE, Sonveaux P, Viollet B, Sakamoto K, Feron O, Horman S, Vanoverschelde JL, Beauloye C, Bertrand L: Role of AMP-activated protein kinase in regulating hypoxic survival and proliferation of mesenchymal stem cells. Cardiovasc Res 2014;101:20-29.
33.
Vermes I, Haanen C, Reutelingsperger C: Flow cytometry of apoptotic cell death. J Immunol Methods 2000;243:167-190.
34.
Kobayashi H, Ouchi N, Kihara S, Walsh K, Kumada M, Abe Y, Funahashi T, Matsuzawa Y: Selective suppression of endothelial cell apoptosis by the high molecular weight form of adiponectin. Circ Res 2004;94:e27-31.
35.
Chandrasekar B, Boylston WH, Venkatachalam K, Webster NJ, Prabhu SD, Valente AJ: Adiponectin blocks interleukin-18-mediated endothelial cell death via APPL1-dependent AMP-activated protein kinase (AMPK) activation and IKK/NF-kappaB/PTEN suppression. J Biol Chem 2008;283:24889-24898.
36.
Iwabu M, Yamauchi T, Okada-Iwabu M, Sato K, Nakagawa T, Funata M, Yamaguchi M, Namiki S, Nakayama R, Tabata M, Ogata H, Kubota N, Takamoto I, Hayashi YK, Yamauchi N, Waki H, Fukayama M, Nishino I, Tokuyama K, Ueki K, Oike Y, Ishii S, Hirose K, Shimizu T, Touhara K, Kadowaki T: Adiponectin and AdipoR1 regulate PGC-1alpha and mitochondria by Ca(2+) and AMPK/SIRT1. Nature 2010;464:1313-1319.
37.
Cory S, Huang DC, Adams JM: The Bcl-2 family: roles in cell survival and oncogenesis. Oncogene 2003;22:8590-8607.
38.
Wu X, Motoshima H, Mahadev K, Stalker TJ, Scalia R, Goldstein BJ: Involvement of AMP-activated protein kinase in glucose uptake stimulated by the globular domain of adiponectin in primary rat adipocytes. Diabetes 2003;52:1355-1363.
39.
Huang PH, Chen JS, Tsai HY, Chen YH, Lin FY, Leu HB, Wu TC, Lin SJ, Chen JW: Globular adiponectin improves high glucose-suppressed endothelial progenitor cell function through endothelial nitric oxide synthase dependent mechanisms. J Mol Cell Cardiol 2011;51:109-119.
40.
Adams V, Heiker JT, Hollriegel R, Beck EB, Woitek FJ, Erbs S, Bluher M, Stumvoll M, Beck-Sickinger AG, Schuler G, Linke A: Adiponectin promotes the migration of circulating angiogenic cells through p38-mediated induction of the CXCR4 receptor. Int J Cardiol 2013;167:2039-2046.
41.
Hou M, Liu J, Liu F, Liu K, Yu B: C1q tumor necrosis factor-related protein-3 protects mesenchymal stem cells against hypoxia- and serum deprivation-induced apoptosis through the phosphoinositide 3-kinase/Akt pathway. Int J Mol Med 2014;33:97-104.
42.
Fotia C, Massa A, Boriani F, Baldini N, Granchi D: Hypoxia enhances proliferation and stemness of human adipose-derived mesenchymal stem cells. Cytotechnology 2015;67:1073-1084.
43.
Lavrentieva A, Majore I, Kasper C, Hass R: Effects of hypoxic culture conditions on umbilical cord-derived human mesenchymal stem cells. Cell Commun Signal 2010;8:18.
44.
Chen CT, Hsu SH, Wei YH: Upregulation of mitochondrial function and antioxidant defense in the differentiation of stem cells. Biochim Biophys Acta 2010;1800:257-263.
45.
Kiani AA, Kazemi A, Halabian R, Mohammadipour M, Jahanian-Najafabadi A, Roudkenar MH: HIF-1alpha confers resistance to induced stress in bone marrow-derived mesenchymal stem cells. Arch Med Res 2013;44:185-193.
46.
Wagegg M, Gaber T, Lohanatha FL, Hahne M, Strehl C, Fangradt M, Tran CL, Schonbeck K, Hoff P, Ode A, Perka C, Duda GN, Buttgereit F: Hypoxia promotes osteogenesis but suppresses adipogenesis of human mesenchymal stromal cells in a hypoxia-inducible factor-1 dependent manner. PLoS One 2012;7:e46483.
47.
Yokota T, Oritani K, Takahashi I, Ishikawa J, Matsuyama A, Ouchi N, Kihara S, Funahashi T, Tenner AJ, Tomiyama Y, Matsuzawa Y: Adiponectin, a new member of the family of soluble defense collagens, negatively regulates the growth of myelomonocytic progenitors and the functions of macrophages. Blood 2000;96:1723-1732.
48.
Kang JH, Lee YY, Yu BY, Yang BS, Cho KH, Yoon DK, Roh YK: Adiponectin induces growth arrest and apoptosis of MDA-MB-231 breast cancer cell. Arch Pharm Res 2005;28:1263-1269.
49.
Zeng F, Shi J, Long Y, Tian H, Li X, Zhao AZ, Li RF, Chen T: Adiponectin and Endometrial Cancer: A Systematic Review and Meta-Analysis. Cell Physiol Biochem 2015;36:1670-1678.
50.
Sun Y, Chen X: Effect of adiponectin on apoptosis: proapoptosis or antiapoptosis? Biofactors 2010;36:179-186.
51.
Yamauchi T, Nio Y, Maki T, Kobayashi M, Takazawa T, Iwabu M, Okada-Iwabu M, Kawamoto S, Kubota N, Kubota T, Ito Y, Kamon J, Tsuchida A, Kumagai K, Kozono H, Hada Y, Ogata H, Tokuyama K, Tsunoda M, Ide T, Murakami K, Awazawa M, Takamoto I, Froguel P, Hara K, Tobe K, Nagai R, Ueki K, Kadowaki T: Targeted disruption of AdipoR1 and AdipoR2 causes abrogation of adiponectin binding and metabolic actions. Nat Med 2007;13:332-339.
52.
Lee HW, Kim SY, Kim AY, Lee EJ, Choi JY, Kim JB: Adiponectin stimulates osteoblast differentiation through induction of COX2 in mesenchymal progenitor cells. Stem Cells 2009;27:2254-2262.
53.
Palanivel R Fang X, Park M, Eguchi M, Pallan S, De Girolamo S, Liu Y, Wang Y, Xu A, Sweeney G: Globular and full-length forms of adiponectin mediate specific changes in glucose and fatty acid uptake and metabolism in cardiomyocytes. Cardiovasc Res 2007;75:148-157.
54.
Yan W, Zhang H, Liu P, Wang H, Liu J, Gao C, Liu Y, Lian K, Yang L, Sun L, Guo Y, Zhang L, Dong L, Lau WB, Gao E, Gao F, Xiong L, Wang H, Qu Y, Tao L: Impaired mitochondrial biogenesis due to dysfunctional adiponectin-AMPK-PGC-1alpha signaling contributing to increased vulnerability in diabetic heart. Basic Res Cardiol 2013;108:329.
55.
Huang B, Cheng X, Wang D, Peng M, Xue Z, Da Y, Zhang N, Yao Z, Li M, Xu A, Zhang R: Adiponectin promotes pancreatic cancer progression by inhibiting apoptosis via the activation of AMPK/Sirt1/PGC-1alpha signaling. Oncotarget 2014;5:4732-4745.
56.
Horman S, Beauloye C, Vanoverschelde JL, Bertrand L: AMP-activated protein kinase in the control of cardiac metabolism and remodeling. Curr Heart Fail Rep 2012;9:164-173.
57.
Gunduz D, Klewer M, Bauer P, Tanislav C, Sedding D, Rohrbach S, Schulz R, Aslam M: Compound C inhibits in vitro angiogenesis and ameliorates thrombin-induced endothelial barrier failure. Eur J Pharmacol 2015;768:165-172.
58.
Liu X, Chhipa RR, Nakano I, Dasgupta B: The AMPK inhibitor compound C is a potent AMPK-independent antiglioma agent. Mol Cancer Ther 2014;13:596-605.
59.
Wang Y, Gao E, Tao L, Lau WB, Yuan Y, Goldstein BJ, Lopez BL, Christopher TA, Tian R, Koch W, Ma XL: AMP-activated protein kinase deficiency enhances myocardial ischemia/reperfusion injury but has minimal effect on the antioxidant/antinitrative protection of adiponectin. Circulation 2009;119:835-844.
60.
Cory S, Adams JM: The Bcl-2 family: regulators of the cellular life-or-death switch. Nat Rev Cancer 2002;2:647-656.
61.
Jung TW, Lee JY, Shim WS, Kang ES, Kim JS, Ahn CW, Lee HC, Cha BS: Adiponectin protects human neuroblastoma SH-SY5Y cells against acetaldehyde-induced cytotoxicity. Biochem Pharmacol 2006;72:616-623.
62.
Gross A, Jockel J, Wei MC, Korsmeyer SJ: Enforced dimerization of BAX results in its translocation, mitochondrial dysfunction and apoptosis. EMBO J 1998;17:3878-3885.
63.
Crompton M: Bax, Bid and the permeabilization of the mitochondrial outer membrane in apoptosis. Curr Opin Cell Biol 2000;12:414-419.
64.
Korytowski W, Basova LV, Pilat A, Kernstock RM, Girotti AW: Permeabilization of the mitochondrial outer membrane by Bax/truncated Bid (tBid) proteins as sensitized by cardiolipin hydroperoxide translocation: mechanistic implications for the intrinsic pathway of oxidative apoptosis. J Biol Chem 2011;286:26334-26343.
65.
Oltvai ZN, Milliman CL, Korsmeyer SJ: Bcl-2 heterodimerizes in vivo with a conserved homolog, Bax, that accelerates programmed cell death. Cell 1993;74:609-619.
66.
Jung TW, Lee JY, Shim WS, Kang ES, Kim JS, Ahn CW, Lee HC, Cha BS: Adiponectin protects human neuroblastoma SH-SY5Y cells against MPP+-induced cytotoxicity. Biochem Biophys Res Commun 2006;343:564-570.
67.
Lee HC, Wei YH: Oxidative stress, mitochondrial DNA mutation, and apoptosis in aging. Exp Biol Med (Maywood) 2007;232:592-606.
68.
Chen J, Baydoun AR, Xu R, Deng L, Liu X, Zhu W, Shi L, Cong X, Hu S, Chen X: Lysophosphatidic acid protects mesenchymal stem cells against hypoxia and serum deprivation-induced apoptosis. Stem Cells 2008;26:135-145.
69.
Liu XB, Jiang J, Gui C, Hu XY, Xiang MX, Wang JA: Angiopoietin-1 protects mesenchymal stem cells against serum deprivation and hypoxia-induced apoptosis through the PI3K/Akt pathway. Acta Pharmacol Sin 2008;29:815-822.
70.
Xu R, Chen J, Cong X, Hu S, Chen X: Lovastatin protects mesenchymal stem cells against hypoxia- and serum deprivation-induced apoptosis by activation of PI3K/Akt and ERK1/2. J Cell Biochem 2008;103:256-269.
71.
Li N, Yang YJ, Cui HH, Zhang Q, Jin C, Qian HY, Dong QT, Zhang H: Tongxinluo decreases apoptosis of mesenchymal stem cells concentration-dependently under hypoxia and serum deprivation conditions through the AMPK/eNOS pathway. J Cardiovasc Pharmacol 2014;63:265-273.
72.
Zhou H, Li D, Shi C, Xin T, Yang J, Zhou Y, Hu S, Tian F, Wang J, Chen Y: Effects of Exendin-4 on bone marrow mesenchymal stem cell proliferation, migration and apoptosis in vitro. Sci Rep 2015;5:12898.
Open Access License / Drug Dosage / Disclaimer
This article is licensed under the Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International License (CC BY-NC-ND). Usage and distribution for commercial purposes as well as any distribution of modified material requires written permission. Drug Dosage: The authors and the publisher have exerted every effort to ensure that drug selection and dosage set forth in this text are in accord with current recommendations and practice at the time of publication. However, in view of ongoing research, changes in government regulations, and the constant flow of information relating to drug therapy and drug reactions, the reader is urged to check the package insert for each drug for any changes in indications and dosage and for added warnings and precautions. This is particularly important when the recommended agent is a new and/or infrequently employed drug. Disclaimer: The statements, opinions and data contained in this publication are solely those of the individual authors and contributors and not of the publishers and the editor(s). The appearance of advertisements or/and product references in the publication is not a warranty, endorsement, or approval of the products or services advertised or of their effectiveness, quality or safety. The publisher and the editor(s) disclaim responsibility for any injury to persons or property resulting from any ideas, methods, instructions or products referred to in the content or advertisements.