Synergistic Activation of LEPR and ADRB2 Induced by Leptin Enhances Reactive Oxygen Specie Generation in Triple-Negative Breast Cancer Cells
Article information
Abstract
Purpose
Leptin interacts not only with leptin receptor (LEPR) but also engages with other receptors. While the pro-oncogenic effects of the adrenergic receptor β2 (ADRB2) are well-established, the role of leptin in activating ADRB2 in triple-negative breast cancer (TNBC) remains unclear.
Materials and Methods
The pro-carcinogenic effects of LEPR were investigated using murine TNBC cell lines, 4T1 and EMT6, and a tumor-bearing mouse model. Expression levels of LEPR, NADPH oxidase 4 (NOX4), and ADRB2 in TNBC cells and tumor tissues were analyzed via western blot and quantitative real-time polymerase chain reaction. Changes in reactive oxygen species (ROS) levels were assessed using flow cytometry and MitoSox staining, while immunofluorescence double-staining confirmed the co-localization of LEPR and ADRB2.
Results
LEPR activation promoted NOX4-derived ROS and mitochondrial ROS production, facilitating TNBC cell proliferation and migration, effects which were mitigated by the LEPR inhibitor Allo-aca. Co-expression of LEPR and ADRB2 was observed on cell membranes, and bioinformatics data revealed a positive correlation between the two receptors. Leptin activated both LEPR and ADRB2, enhancing intracellular ROS generation and promoting tumor progression, which was effectively countered by a specific ADRB2 inhibitor ICI118551. In vivo, leptin injection accelerated tumor growth and lung metastases without affecting appetite, while treatments with Allo-aca or ICI118551 mitigated these effects.
Conclusion
This study demonstrates that leptin stimulates the growth and metastasis of TNBC through the activation of both LEPR and ADRB2, resulting in increased ROS production. These findings highlight LEPR and ADRB2 as potential biomarkers and therapeutic targets in TNBC.
Introduction
Breast cancer development is influenced by a complex interplay of genetic, hormonal, and environmental factors. Obesity is associated with an elevated risk of developing breast cancer, particularly in postmenopausal women, and is linked to a poorer disease outcome for women of all ages [1]. Observational studies suggest that exercise combined with weight loss may improve breast cancer outcomes [2]. Given the presence of adipose tissue in the mammary gland, it is plausible to hypothesize an interaction between adipocytes and the substances they release, including leptin, and the progression of the tumor.
Leptin, primarily produced by mature adipocytes, plays a crucial role in regulating food intake, body weight, and maintaining energy homeostasis [3]. Individuals with obesity exhibit higher leptin levels compared to lean counterparts [4]. The leptin receptor (LEPR), also known as Ob-R, is a transmembrane receptor with a helical structure, related to class I cytokine receptors [5]. Ubiquitously expressed in various tissues including the pancreas, placenta, adrenal glands, liver, and breast cells [6], both leptin and LEPR are overexpressed in breast cancer epithelial cells when compared to non-cancerous mammary epithelial cells [7]. This difference in LEPR expression holds significance as a potential prognostic and/or predictive biomarker [7]. Notably, recent research identified LEPR as a predictive factor for pathological complete response in early breast tumors managed with preoperative chemotherapy, independent of the molecular subtype [8].
In breast cancer tumorigenesis, leptin appears to be a key driver. An increasing body of evidence demonstrates that leptin enhances cell growth in both normal and malignant breast epithelial cells by activating various signaling pathways [6,9]. Animal studies using mice with genetically leptin-deficient Lepob/ob or LEPR-deficient LEPRdb/db did not detect mammary tumors in either case [10], emphasizing the essential role of an intact leptin axis in tumor development. However, the specific mechanism by which leptin/LEPR axis affects triple-negative breast cancer (TNBC) progression remains unclear.
In the dysregulation of energy balance in obesity, leptin, insulin, and catecholamine resistance have been characterized [11]. Particularly, catecholamine resistance is a key feature of the obese and may even predict future weight gain in some patients [12]. Emerging studies reveal a close connection between leptin and the sympathoadrenal axis. In adipose tissues, the lipolytic effect of leptin is mediated through the action of sympathetic nerve fibers that innervate the adipose tissue [13]. Leptin treatment leads to an increase in norepinephrine (NE) levels in adipose tissue. Interestingly, while NE levels in white adipose tissues dissected from leptin-treated animals are significantly higher than those in controls, leptin treatment does not affect serum NE levels, indicating a localized increase in NE release in white fat, but not systemically [13]. Additionally, in bone metabolism, leptin influences bone formation through both direct and indirect pathways. It stimulates the sympathetic nervous system (SNS) in the central medial hypothalamic nucleus, with released NE to activate adrenergic receptor β2 (ADRB2) on osteoblasts, indirectly impeding bone formation. In the periphery, leptin directly affects bone formation by interacting with LEPR on osteoblasts [14].
Collectively, these studies underscore a robust association between leptin and the SNS/NE/ADRB axis. In the mammary gland, rich in adipocytes and abundantly innervated by SNS nerve terminals [15], the pro-carcinogenic effect of ADRB2 in TNBC progression and the tumor microenvironment is well-established in our previous research [16] and others [17]. Yet, it remains unknown whether leptin affects the SNS/ADRB2 receptor axis in breast cancer cells, and the collaborative mechanism promoting breast cancer progression has not been reported.
Building on these insights, we elucidated the promotional effect of leptin on breast cancer growth and metastasis in vitro and in vivo. Importantly, we discovered that leptin activates both LEPR and ADRB2 co-expressed on TNBC cells, exerting a pro-carcinogenic effect by promoting an increase in reactive oxygen species (ROS) generation. The application of the LEPR inhibitor Allo-aca and the ADRB2-specific inhibitor ICI118551 significantly diminished tumor size in the mouse model. Our study offers novel perspectives for TNBC treatment, emphasizing the potential of co-targeting LEPR/ADRB2 as predictive and therapeutic focal points.
Materials and Methods
1. Cell culture and treatment
Two murine-derived TNBC cell lines, 4T1 and EMT6 (purchased from ATCC), were cultured in RPMI-1640 medium (BI, Israel, South America) supplemented with 10% fetal bovine serum (Newzerum) under standard conditions of 37°C and 5% CO2. Cells were seeded in six-well plates and allowed to reach approximately 60% confluence before being subjected to treatment.
For experimentation, cells were treated with varying concentrations (10, 50, 100, 200 ng/mL) of recombinant mouse leptin (R&D Systems) for a duration of 24 hours. Additionally, the LEPR antagonist, Allo-aca (100 nM for 24 hours, MCE), and the specific ADRB2 inhibitor, ICI118551 (10 μM for 24 hours, Sigma-Aldrich), were separately added to 4T1 and EMT6 cells at appropriate densities prior to further experimentation.
2. Quantitative Real-time PCR
Total RNA was extracted from the samples, followed by cDNA synthesis using the Hifair II 1st Strand cDNA Synthesis Kit (Yeasen). Subsequently, DNA amplification was carried out using the UltraSYBR Mixture (Cwbio). The relative expression levels of target genes were determined using the formula 2-ΔΔCT, with normalization to β-actin.
The primer sequences utilized were as follows: LEPR: forward 5′-AGCTAGGTGTAAACTGGGACA-3′, reverse 5′-GCAGAGGCGAATCATCTATGAC-3′; ADRB2: forward 5′-ATGTCGGTTATCGTCCTGGC-3′, reverse 5′-GGTTTGTAGTCGCTCGAACTTG-3′; NADPH oxidase 4 (NOX4): forward 5′-TTTCTCAGGTGTGCATGTAGC-3′, reverse 5′-GCGTAGGTAGAAGCTGTAACCA-3′; β-actin: forward 5′-GGCTGTATTCCCCTCCATCG-3′, reverse 5′-CCAGTTGGTAACAATGCCATGT-3′.
3. Western blot
Total protein was extracted utilizing RIPA lysis buffer (Solarbio), followed by quantification using the BCA assay kit (Beyotime Biotechnology). Subsequently, protein samples were denatured in a metal bath at 95°C for 8 minutes. Electrophoresis was then performed using a 10% sodium dodecyl sulfate polyacrylamide gel electrophoresis gel, and proteins were transferred onto a polyvinylidene difluoride membrane (Millipore). The membrane was incubated with 10% skimmed milk at room temperature for 1 hour, followed by overnight incubation at 4°C with primary antibodies. Anti-LEPR and anti-ADRB2 antibodies were sourced from Abcam (ab5593, ab182136), anti-NOX4 antibody from Boster (A00403), and anti-β-actin antibody from Santa Cruz Biotechnology (sc-81178, Santa Cruz Biotechnology). After washing thrice with PBST (phosphate buffered saline with Tween 20), membranes were incubated with secondary antibodies at room temperature for 1 hour. Protein bands were visualized using chemiluminescent horseradish peroxidase substrate (ECL, WBKlS0100, Millipore), and band densities were analyzed using Image J.
4. Cell viability assay
Cell viability was assessed using the Cell Counting Kit-8 (CCK-8; Beyotime Biotechnology) following the manufacturer’s instructions. Briefly, cells were seeded in 96-well plates and allowed to adhere for 24 hours before treatment with varying concentrations of leptin (10, 50, 100, and 200 ng/mL) for an additional 24 hours. In a separate experiment, cells were treated with leptin (100 ng/mL) alone or in combination with Allo-aca (100 nM). Following treatment, CCK-8 solution was added to each well, and absorbance was measured at 450 nm to evaluate cell viability.
5. Clone formation assay
EMT6 single-cell suspension were seeded in a six-well plate at a density of 500 cells per well. Once adhered, cells were subjected to treatment with either leptin or Allo-aca, and the culture medium was refreshed every three days. The experiment was concluded when the number of cells in most individual clones exceeded 50 cells, as determined under microscopic observation. Following this, cells were fixed with 4% paraformaldehyde and stained with 1 mL of crystal violet dye for 10 minutes per well. After rinsing with PBS, the cells were allowed to air-dry before being photographed.
6. Wound closure assay
EMT6 cells were seeded in a six-well plate at a density of 2×105 cells and allowed to incubate overnight. Using a pipette tip, scratches were carefully made on the cell monolayer. Following this, the cells were gently washed three times with PBS and treated with either leptin or Allo-aca for 18 hours. The scratches were observed and photographed under a microscope and the rate of scratch closure was quantitatively analyzed using ImageJ software.
7. Transwell migration assay
After pre-culturing 4T1 cells in serum-free medium for 24 hours, a single-cell suspension was prepared at a density of 5×105 cells/mL. Subsequently, 200 μL of the cell suspension was added to the upper chamber of a transwell chamber. In the lower chamber of the 24-well plate, 600 μL of medium containing 15% fetal bovine serum along with either leptin or Allo-aca was added. The cells were then incubated at 37°C for 24 hours. Then the transwell chamber was washed twice with PBS, fixed with methanol, and stained with 0.1% crystal violet. The upper layer of non-migrated cells was gently wiped off with a cotton swab. Migrated cells were then photographed and quantitatively analyzed using Image J.
8. ROS measurement
The levels of ROS were assessed using the ROS fluorescent probe DCFH-DA (Invitrogen) via flow cytometry. Following 24 hours of various treatments, cells were harvested, washed with PBS, and then incubated with serum-free medium containing 10 μM DCFH-DA for 20 minutes, protected from light. The levels of ROS were measured using flow cytometry (BD FACSCanto II).
9. Measurement of mitochondrial ROS
4T1 cells and EMT6 cells were treated with MitoSOX Red fluorescent dye (5 μM, Thermo Fisher, Waltham, MA) and incubated at 37°C for 30 minutes. Following the incubation period, cells were washed with PBS and examined under a confocal microscope (Olympus FV1000).
10. Double labeling immunofluorescence analysis
After treatment with leptin (100 ng/mL) alone or in combination with Allo-aca (100 nM) for 24 hours, cells were washed with PBS and fixed with 4% paraformaldehyde at room temperature. The cells were blocked with 5% goat serum for 30 minutes and then incubated overnight at 4°C with a 1:100 dilution of anti-LEPR antibody (sc-8391, Santa Cruz Biotechnology) and a 1:200 dilution of anti-ADRB2 antibody (ab128136, Abcam). This was followed by incubation with the corresponding Alexa Fluor-conjugated secondary antibodies (1:200, Affinity) for 1 hour in the dark. Cell nuclei were stained with DAPI. Immunoreactivity was captured using a confocal microscope with 488 and 594 laser excitations (Olympus FV1000), and the fluorescence intensity was analyzed using the coloc2 plug-in (ver. 2.0, Fiji) of Image J v1.8.0.
11. In vivo tumorigenesis
Female BALB/c mice, aged 6-8 weeks, were procured from Vital River Lab Animal Technology Co., Ltd. All experimental procedures were ethically approved by the Medical Ethics Committee, Nankai University (2023-SYDWLL-000531). Following subcutaneous inoculation of 1×105 4T1 cells into the breast gland of the mice, the animals were randomly divided into four groups: control group, leptin group (intraperitoneal injection, 1 mg/kg), leptin (1 mg/kg) combined with Allo-aca (subcutaneous injection, 0.1 mg/kg), and leptin (1 mg/kg) combined with ICI118551 (intraperitoneal injection, 0.5 mg/kg). Each drug combination was administered once every two days. Throughout the experiment, the mice’s food intake and body weight were monitored and recorded every two days. Tumor volume was measured and calculated using the formula V=(length×width2)/2. Following 3 weeks of drug administration, the mice were euthanized, and blood serum from the eyeball, as well as tumors and lungs, were collected for subsequent experiments.
12. Glutathione assay
The measurement of glutathione (GSH) content in serum or tumor samples was conducted using a GSH kit (NjJcBio) following the manufacturer’s instructions. Absorbance readings were taken at a wavelength of 420 nm.
13. Lung metastasis in tumor-implanting mice
The mouse lung tissue sections were stained with hematoxylin and eosin (H&E) and imaged at Zhongke Guanghua Biotechnology Company.
14. Bioinformation analysis
Survival analysis data was obtained from the Kaplan-Meier plotter database (http://kmplot.com/analysis/index.php?p=service&cancer), using the ‘auto-select best cutoff’ option for cutoff determination. Correlation analysis was performed using bc-GenExMiner (http://bcgenex.ico.unicancer.f/BC-GEM/GEM-requete.php) and GEPIA (http://gepia.cancer-pku.cn/). Additionally, the prognostic significance in breast cancer patients was assessed by receiver operating characteristic (ROC) curve using the Plotter platform (http://rocplt.org).
15. Statistical analysis
The data was presented as mean±standard error of mean, and statistical analysis was performed using Prism 9.0 (GraphPad). Student’s t test was utilized for comparisons between two groups, while one-way ANOVA followed by Tukey’s test was employed for comparisons among multiple groups. Statistical significance was set at p < 0.05.
Results
1. Leptin upregulates LEPR expression in TNBC cells
Using the publicly accessible Kaplan-Meier plotter database, we identified that elevated LEPR expression in TNBC tissues correlated with poorer overall survival (OS) and distant metastasis-free survival (p < 0.05) (Fig. 1A and B). We further investigated the impact of leptin on LEPR expression in murine TNBC cell lines, 4T1 and EMT6. Both cell types showed increased LEPR expression in response to leptin treatment (0, 10, 50, 100, and 200 ng/mL) for 24 hours, with peak expression at 100 ng/mL (Fig. 1C and D). Therefore, we used 100 ng/mL leptin for subsequent experiments. To counteract leptin’s effects, we used the LEPR antagonist peptide Allo-aca (Allo). Allo alone did not significantly alter LEPR mRNA and protein levels in 4T1 or EMT6 cells. However, it effectively inhibited leptin-induced LEPR upregulation (p < 0.05) (Fig. 1E and F). These findings suggest that LEPR in murine TNBC cell lines can be activated by leptin, and Allo may suppress this effect.
2. LEPR activation promotes TNBC cell proliferation and migration
To investigate the role of LEPR in breast cancer progression, we examined cell proliferation using CCK-8 and colony formation assays. Treatment of 4T1 and EMT6 cells with varying concentrations of leptin for 24 hours increased cell viability, peaking at 100 ng/mL for 4T1 cells and 50 ng/mL for EMT6 cells (p < 0.05) (Fig. 2A). While Allo alone did not significantly affect cell proliferation, it counteracted the proliferative effects of 100 ng/ml leptin (p < 0.05) (Fig. 2B). Colony formation assays showed similar results in EMT6 cells (p < 0.05) (Fig. 2C).
We also explored the influence of the leptin/LEPR axis on TNBC cell migration. In both the wound closure assay for EMT6 cells and the transwell migration assay for 4T1 cells, leptin increased cell migration, while Allo significantly reduced the stimulatory effects of leptin (p < 0.05) (Fig. 2D and E).
3. Leptin induces NOX4-derived ROS and mitochondrial ROS production in TNBC cells
ROS production is crucial in both obesity and cancer. Leptin has been shown to induce ROS in various tissues, suggesting a potential signaling pathway via LEPR. In mammary gland cells, leptin induces ROS through NOX5 [18], but its effects on ROS in TNBC cells 4T1 and EMT6 were unexplored. We evaluated ROS production using the DCFH-DA probe, finding significantly elevated ROS levels in both cell types after leptin stimulation, although not in a dose-dependent manner (Fig. 3A). Allo alone did not affect ROS levels but inhibited leptin-induced ROS production (p < 0.05) (Fig. 3B).
The primary drivers of cellular ROS production in physiological and pathological processes are NADPH oxidases (NOXs), with mitochondrial ROS serving as another significant source (Fig. 3C). Among NOX family members, NOX4 exhibited high expression in both 4T1 and EMT6 cells (data not shown). Leptin treatment increased NOX4 mRNA (p < 0.05) (Fig. 3D) and protein expression (p < 0.05) (Fig. 3E) in both cell lines, which was significantly reduced by Allo (p < 0.05) (Fig. 3F and G).Using MitoSOX Red, we found that leptin-induced mitochondrial ROS (mtROS) production was also significantly reduced by Allo treatment (Fig. 3H and I). Leptin treatment induced ROS generation, which contributed to the proliferation of EMT6 cells as shown by cell viability analysis, and promoted 4T1 cell migration as evidenced by wound closure assays. However, the ROS scavengers N-acetylcysteine or superoxide dismutase were able to eliminate the leptin-induced ROS effect (S1 Fig.). These findings suggest that the leptin/LEPR axis facilitates TNBC cell proliferation and migration by increasing NOX4-derived ROS and mtROS production, while Allo suppresses this effect.
4. Leptin upregulates ADRB2 expression in TNBC cells
To probe into the potential association between leptin and ADRB2 in breast cancer cells, we used quantitative real-time PCR and protein blotting to measure ADRB2 expression in the presence of leptin. Our results showed a significant increase in ADRB2 mRNA and protein levels in leptin-stimulated 4T1 and EMT6 cells (p < 0.05) (Fig. 4A and B). Furthermore, Allo, an inhibitor of LEPR, effectively suppressed leptin-induced upregulation of ADRB2 expression (p < 0.05) (Fig. 4C and D). Immunofluorescence double staining confirmed the presence of both LEPR, conjugated with Alexa Fluor 488, and ADRB2, conjugated with Alexa Fluor 594, on the cell membrane of EMT6 cells (Fig. 4E). Statistical analysis indicated that leptin treatment promoted the co-localization of LEPR and ADRB2, whereas Allo mitigated this effect. Additionally, the ADRB2 antagonist ICI118551 inhibited the co-localization of these receptors (Fig. 4F-J). The Pearson correlation coefficient statistics plot reconfirmed the fluorescence intensity changes of LEPR and ADRB2 in EMT6 cells under different treatments (Fig. 4K). These findings suggest a potential link between LEPR and ADRB2 in TNBC, like conditions observed in obesity. Moreover, they indicate that leptin not only activates LEPR but also induces the upregulation of ADRB2.
5. ADRB2 mediates leptin-induced ROS generation in TNBC cells
To determine if ADRB2 plays a role in leptin-induced ROS generation, we conducted flow cytometry analysis. Our findings revealed that ICI118551 effectively inhibited leptin-induced ROS generation in both 4T1 and EMT6 cells (Fig. 5A). We then examined changes in ADRB2 and NOX4 expression in leptin-stimulated 4T1 cells treated with ICI118551 using immunoblotting. The results showed that ICI118551 treatment led to a downregulation of both ADRB2 and NOX4 expression, which were enhanced by leptin (p < 0.05) (Fig. 5B). We also assessed mtROS levels through confocal microscopy. The analysis demonstrated that mtROS levels elevated in the presence of leptin compared to the control group, and this increase was effectively inhibited by ICI118551 treatment (Fig. 5C). Collectively, our data indicate that leptin increases ROS levels by concurrently activating both LEPR and ADRB2 receptors, thereby fostering the progression of TNBC.
6. LEPR and ADRB2 in leptin-promoted breast cancer progression in vivo
To further validate our findings, we employed a transplanted tumor-bearing mouse model by inoculating 4T1 cells for in vivo experiments. Seven days post-cell inoculation, BALB/c mice were randomly assigned to receive one of the following treatments: normal saline, leptin, leptin combined with Allo, and leptin combined with ICI118551. Drug injections continued for 3 weeks, during which we monitored diet consumption, body weight, and tumor volume (Fig. 6A). Compared to the control group, the leptin group showed significantly larger tumor volumes and heavier tumor weights (p < 0.05) (Fig. 6B and C). However, Co-administration with the LEPR antagonist Allo or the ADRB2 inhibitor ICI118551 inhibited tumor growth compared to the leptin-alone group (p < 0.05) (Fig. 6B and C).
To ensure leptin’s effects on breast cancer progression were not indirectly due to changes in diet and energy metabolism, we monitored food consumption and body weight. Drug administration had little effect on appetite (Fig. 6D). Mice in the leptin group lost a significant weight compared to the control group, with weight reduction peaking around the 10th day post-leptin injection. This weight loss stabilized over time and was alleviated by combined treatment with Allo or ICI118551 (Fig. 6E). H&E staining of lung tissue sections showed significantly larger metastatic nodules in the leptin group, while combined treatment with Allo or ICI118551 reduced nodule size (Fig. 6F).
GSH, a crucial antioxidant in redox homeostasis of tumor cells, was significantly reduced in the leptin group compared to controls, but this reduction was reversed by combination treatment with Allo or ICI118551 (p < 0.05) (Fig. 6G). Lastly, we quantified mRNA and protein levels in mouse tumor tissues. Consistent with our in vitro results, mRNA and protein expression of LEPR, ADRB2, and NOX4 were significantly upregulated in the leptin group compared to controls. This upregulation was reversed by combined treatment with Allo or ICI118551 (p < 0.05) (Fig. 6H and I).
7. The clinical significance of LEPR and ADRB2 in breast cancer patients
ROC curve analysis is a method used to evaluate the performance of diagnostic classification methods, with the area under the curve (AUC) serving as a key metric. In our study, both LEPR and ADRB2 had an AUC value of 0.629, indicating some diagnostic predictive ability (p < 0.001) (Fig. 7A and B), thus suggesting their potential as diagnostic markers of breast cancer. We further analyzed the correlation between LEPR and ADRB2 using the bc-GenExMiner and GEPIA databases. The data revealed a positive correlation between LEPR and both ADRB2 and NOX4, with ADRB2 showing a stronger positive trend (Fig. 7C-E). In contrast, glutathione peroxidase 4, a ROS scavenging enzyme, exhibited a negative correlation with LEPR (Fig. 7C). Kaplan-Meier plotter analysis demonstrated that high expression of LEPR and ADRB2 was associated with poor relapse-free survival in TNBC patients (p=0.007) (Fig. 7F).
In summary, our study, conducted through both in vitro and in vivo experiments, demonstrates that concurrent activation of the membrane receptors LEPR and ADRB2 on TNBC cells stimulates NOX4-mediated cellular ROS generation and mitochondrial ROS production, fostering the proliferation and metastasis of TNBC (Fig. 7G). These findings suggest that dual targeting of LEPR and ADRB2 could be a promising predictive and therapeutic strategy in the clinical management of TNBC patients.
Discussion
Leptin, known for its role in regulating energy balance and produced mainly by adipose tissue, has been implicated in promoting breast cancer progression through various pathways beyond its own receptor, LEPR [19]. Our study reveals that leptin not only activates LEPR but also ADRB2 receptors in TNBC cells and mouse models, leading to increased tumor growth and metastasis through the generation of ROS.
Research have reported that leptin interacts with key signaling pathways such as epidermal growth factor receptor (EGFR), and human epidermal growth factor receptor 2 (HER2) [20]. Notably, leptin could transactivate HER2 through EGFR and JAK2 activation, further promoting breast cancer progression [21,22]. Additionally, the role of ADRB2 receptors in tumor progression has been highlighted, particularly in gastric, ovarian, and breast cancers. Evidence has shown adrenergic nerve innervation in the tumor microenvironment and high expression of ADRB2 in tumor tissues [15,23,24]. While leptin’s role in adipose tissue includes lipolytic effects mediated by the SNS, studies have yet to establish a connection between leptin and the adrenergic ADRB2 receptor in breast cancer.
New findings from this study reveal that leptin can activate both LEPR and ADRB2 in TNBC cells and in 4T1 tumor-bearing mice. However, there are limitations to our animal experiments, as results from mouse models may not be directly applicable to patients. Nude mouse models have limitations due to their T-cell–deficient environment. Therefore, we used the BALB/c 4T1 mouse model, which is reliable and well-established in our lab.
Obesity-related hyperleptinemia is a recognized risk factor for breast cancer, with leptin and LEPR exhibiting heightened expression in breast cancer tissues compared to non-cancerous tissue [25]. Immunohistochemical analyses further revealed an upregulation of LEPR across various stages of breast cancer, from primary tumors to metastases [26]. Leptin acts as a growth factor, influencing tumor initiation, proliferation, invasion, migration, and angiogenesis [27]. Additionally, leptin’s pro-inflammatory effects may potentially enhance the efficacy of immune checkpoint inhibitors [28], though this area remains controversial.
Leptin is primarily synthesized and released into circulation by adipose tissue. It is also produced in smaller amount by various other tissues such as skeletal muscle, the brain, ovaries, immune cells, and both normal and malignant breast tissue [29,30]. As a mediator of long-term energy balance regulation, leptin plays a crucial role in suppressing food intake and inducing weight loss. Our study shows that administering leptin injections leads to weight loss in tumor-bearing mice without affecting food intake, consistent with leptin’s role in energy balance regulation. Moreover, combining long-term exercise with controlled caloric intake can effectively reduce leptin synthesis and secretion [31].
The soluble LEPR (sOB-R) plays a crucial role in inhibiting leptin’s effect. As a structural analog of the cell membrane LEPR, sOB-R competes with it for leptin binding. When leptin binds to sOB-R, its bioavailability decreases because it cannot bind to cell membrane LEPR [32]. Therefore, stimulating sOB-R production may inhibit leptin’s bioactivity and reduce oxidative stress. Compounds like fobol esters and palmitic acid have been found to stimulate sOB-R production, thus inhibiting leptin [33].
ROS plays a dual role in tumor progression, acting both as pro-oncogenic and anti-oncogenic agents [34]. Our study aligns with previous research showing that ROS accumulation promotes TNBC progression by activating diverse downstream signaling pathways [35-37]. Specifically, inhibitors targeting LEPR (Allo) and ADRB2 (ICI118551) effectively suppressed ROS generation in TNBC cells, inhibiting tumor growth. However, our study has limitations, particularly in the detailed exploration of the molecular mechanisms by which LEPR and ADRB2 influence ROS generation. Some studies suggest that LEPR and ADRB2 may regulate ROS production through the JAK2 and ERK signaling pathways. For instance, LEPR activation has been shown to enhance adipogenesis via JAK2/stat3 signaling in bone marrow stromal cells [38]. Similarly, high ADRB2 expression could stimulate JAK2 phosphorylation [39]. In the hypothalamus, ERK mediates leptin’s effects on appetite and thermogenesis [40]. Additionally, NE, an ADRB2 agonist, promotes ERK phosphorylation in breast cancer cells [41]. These findings suggest that the JAK2 and ERK pathways might be important for ROS generation mediated by LEPR and ADRB2, warranting further investigation to elucidate these mechanisms in TNBC.
In conclusion, our study demonstrates that leptin stimulates the growth and metastasis of TNBC, both in vitro and in vivo, through the activation of both LEPR and ADRB2, resulting in increased ROS production. This underscores the importance of identifying LEPR and ADRB2 as novel biomarkers and potential therapeutic targets in TNBC.
Electronic Supplementary Material
Supplementary materials are available at Cancer Research and Treatment website (https://www.e-crt.org).
Notes
Ethical Statement
All experimental procedures were ethically approved by the Medical Ethics Committee, Nankai University (2023-SYDWLL-000531).
Author Contributions
Conceived and designed the analysis: Wang Y, Qin J.
Collected the data: Liu C, Yu J, Du Y.
Contributed data or analysis tools:Xie Y, Song X, Liu C, Yan Y.
Performed the analysis: Liu C, Yu J, Du Y.
Wrote the paper: Liu C, Wang Y, Qin J.
Conflicts of Interest
Conflict of interest relevant to this article was not reported.
Funding
This research was supported by the National Natural Science Foundation of China (31770968, to Yue Wang; 31800661, to Junfang Qin) and the Science Fund Project of Tianjin (No. 21JCYBJC00240 and 20JCYBJC01130).