Skip to main content
Bioactive Materials logoLink to Bioactive Materials
. 2025 Mar 15;49:378–398. doi: 10.1016/j.bioactmat.2025.03.007

Silicified curcumin microspheres Combats cardiovascular diseases via Nrf2/HO-1 pathway

Tianwang Guan a,b,1, Zhenxing Lu b,c,1, Rundong Tai b,c,d,1, Shuai Guo c,1, Zhaowenbin Zhang e,f,⁎⁎, Shaohui Deng c, Jujian Ye b,c,d, Kaiyi Chi g, Binghua Zhang h, Huiwan Chen b,c,d, Zhilin Deng c, Yushen Ke b,c,d, Andong Huang h, Peier Chen b,c,2, Chunming Wang i,j,, Caiwen Ou b,c,2
PMCID: PMC11937612  PMID: 40144796

Abstract

Diabetes and chemotherapy frequently give rise to severe cardiovascular complications, including chemotherapy-induced cardiotoxicity and diabetes-associated vascular remodeling. Nevertheless, the precise epidemiological features of these cardiovascular ailments remain incompletely elucidated, resulting in a dearth of effective therapeutic strategies in clinical settings. To tackle this intricate challenge, we have delved extensively into database resources, conducted comprehensive analyses of pertinent epidemiological data, and designed silicified curcumin (Si/Cur) microspheres as a novel therapeutic approach for cardiovascular diseases. By harnessing the alkaline microenvironment generated by silicon (Si), Si/Cur markedly elevates the bioavailability of curcumin (Cur). Further investigations have elucidated that Si/Cur exerts its therapeutic actions primarily via the Nrf2/HO-1 signaling pathway, effectively suppressing vascular remodeling and mitigating myocardial injury, thus disrupting the vicious cycle of persistent cardiovascular damage. In conclusion, this study integrates clinical cohort research to dissect epidemiological characteristics, directs the design and application of biomaterials, and paves the way for a novel and efficacious therapeutic avenue for the management of cardiovascular diseases.

Keywords: Silicified curcumin, Doxorubicin, Cardiotoxicity, Diabetes, Nrf2/HO-1 signaling pathway, Vascular remodeling, Cardio-oncology

Graphical abstract

Targeting the complex disease cohort of diabetic cancer patients undergoing chemotherapy, silicified curcumin (Si/Cur) has been developed leveraging sophisticated data mining techniques. This material effectively regulates the Nrf2/HO-1 signaling pathway, achieving inhibition of vascular remodeling and alleviation of myocardial injury, thereby potently disrupting the vicious cycle of continuous cardiovascular damage in these coexisting conditions. Created in BioRender. Guan, T. (2024) https://BioRender.com/s59v201.

Image 1

Highlights

  • Siliconized curcumin creates an alkaline environment, enhancing Cur delivery and activity.

  • Siliconized curcumin protected neonatal rat cardiomyocytes from DOX-induced damage.

  • Siliconized curcumin reduces inflammation and oxidative stress under high glucose.

  • Innovative biomaterial design strategies are provided.

1. Introduction

Globally, an astonishing 537 million people are living with diabetes, accounting for 10.5 % of the world's population [1]. Concurrently, a staggering 20 million new cancer cases are diagnosed each year worldwide [2]. These patient populations are particularly susceptible to cardiovascular diseases. For instance, recent research has revealed that chemotherapy drugs can induce cardiotoxicity, leading to heart failure, arrhythmias, and ischemic heart disease [3,4]. Diabetes, on the other hand, triggers vascular remodeling, a condition that remains inadequately controlled by current antidiabetic medications [5,6]. Although various individual treatment strategies currently exist for cardiotoxicity of anticancer treatments and vascular remodeling diseases, such as intramyocardial injection of platelet gel, percutaneous endocardial injection for delivering allogeneic cardiac spheres, and integrating hypoxia-targeting and photothermal therapeutic agents to achieve hypoxia-targeted cancer treatment, thereby reducing the side effects of chemotherapy drugs [[7], [8], [9], [10], [11], [12]], the cardiovascular risks for diabetic patients significantly increase when they develop cancer and undergo chemotherapy. Unfortunately, effective treatments for cardiovascular complications caused by diabetes and chemotherapy are currently lacking. This underscores the urgent need to explore specialized treatment strategies tailored to these patients, aimed at reducing mortality and enhancing their quality of life.

The cardiovascular disease conditions observed in both diabetic patients and cancer patients undergoing chemotherapy are exceedingly complex and influenced by a multitude of factors. In diabetic patients, environmental factors, such as elevated blood sugar levels, stimulate endothelial cells to release a substantial number of inflammatory factors. This promotes the adhesion and infiltration of inflammatory cells into the vascular wall, triggering inflammatory responses in other vascular wall cells, ultimately culminating in vascular remodeling [[13], [14], [15]]. For cancer patients receiving chemotherapy, chemotherapeutic agents directly target cardiomyocytes, inducing widespread cell apoptosis and subsequently leading to cardiac dysfunction [16,17]. Given the intimate relationship between the vascular network and the heart, the coexistence of these two conditions is likely to initiate a vicious cycle of persistent cardiovascular damage. However, owing to the absence of specific epidemiological data on the concomitant occurrence of these conditions, an effective treatment regimen for this type of cardiovascular disease remains elusive. Consequently, it is of paramount importance to conduct comprehensive analyses of such cases through epidemiological cohort studies to identify more efficacious treatment strategies [18].

The precise regulation of physiological processes and the effective treatment of various diseases by natural products from herbal medicines underpin our specific focus on traditional Chinese medicines during the drug screening process [19]. In the present study, utilizing cohort studies and network pharmacology analysis, we identified curcumin (Cur), a natural product derived from herbs, as a potential therapeutic agent for cardiovascular diseases in diabetic patients and cancer patients undergoing chemotherapy. A plethora of studies have established that curcumin possesses potent antioxidant and anti-inflammatory properties [[20], [21], [22]]. Nevertheless, the limited solubility of curcumin in water significantly restricts its application [23]. Following oral administration, over 75 % of curcumin remains unabsorbed and is directly excreted through the intestine. To enhance its solubility, attempts have been made to employ organic solvents, such as dimethyl sulfoxide and ethanol [24], or to establish an alkaline environment [25,26]. However, these methods are unsuitable for biological applications due to the toxicity associated with organic solvents or the corrosiveness of chemical bases, such as sodium hydroxide and potassium hydroxide. Silicon (Si) has attracted considerable attention in the medical field owing to its excellent biocompatibility, as it aids in restoring the normal physiological conditions of crucial vascular cells and cardiomyocytes, thereby effectively ameliorating tissue ischemia and myocardial infarction [27]. Notably, Si ions possess the ability to create a biologically active alkaline environment [[28], [29], [30]]. Based on this principle, we propose leveraging biologically active Si ions to markedly enhance the solubility of curcumin in water, thus offering a promising novel therapy for cardiovascular diseases in diabetic patients and cancer patients undergoing chemotherapy.

Nanomaterials, as carriers, exhibit great potential in the diagnosis and treatment of malignant tumors, as well as in cardiac protection [31,32]. Therefore, we designed silicon (Si) and curcumin (Cur) in the form of nanomaterials, aiming to more effectively deliver Cur to the lesion area. Based on this, we successfully developed a silicon-curcumin (Si/Cur) composite microsphere, which facilitates the long-term delivery of Si/Cur. Preliminary validation results demonstrate its significant therapeutic effects in mouse models of diabetes-related vascular remodeling and doxorubicin-induced cardiotoxicity. To achieve this objective, we have developed a silicified curcumin (Si/Cur) composite microsphere to facilitate the long-term delivery of Si/Cur and have preliminarily validated its therapeutic efficacy in mouse models of diabetes-related vascular remodeling and doxorubicin-induced cardiotoxicity. To further substantiate the therapeutic effect of silicon-curcumin in the composite microspheres, we prepared a soluble Si/Cur solution and validated its efficacy. Additionally, we conducted in-depth investigations using human umbilical vein endothelial cells (HUVECs) and primary cardiomyocytes to elucidate the biological mechanisms by which Si/Cur in the composite material restores cells to their normal physiological state.

2. Results

2.1. Cohort study on the prevalence, prognosis, and cardiovascular damage in diabetic cancer patients undergoing chemotherapy

Selected from the UK Biobank (UKB), 4757 cancer patients who received chemotherapy were included in this prospective cohort study (Table S1). The median follow-up period was 13.3 years [interquartile range (IQR) 12.5–14.1]. The study found that the prevalence of diabetes among cancer patients receiving chemotherapy increased from 2.37 % to 10.92 % between 2006 and 2022 (Fig. 1A). Kaplan-Meier survival analysis revealed that diabetic cancer patients undergoing chemotherapy (DMCA) faced significantly higher risks of all-cause mortality (Fig. 1B) and cardiovascular disease (CVD) mortality (Fig. 1C) compared to cancer patients without diabetes (CA) (both P < 0.001). To minimize false-positive results [33,34], the multivariate model produced robust results after adjusting for confounding factors, showing higher risks of all-cause mortality (Model 1: HR = 1.26, [95 % CI, 1.06–1.49]; Model 2: HR = 1.12, [95 % CI, 1.04–1.21]) (Fig. 1D) and CVD mortality (Model 1: HR = 2.62, [95 % CI, 1.30–5.30]; Model 2: HR = 2.17, [95 % CI, 1.05–4.48]) (Fig. 1E). Fig. 1F demonstrates that CVD (33.72 %) was the leading cause of non-cancer mortality among DMCA patients receiving chemotherapy. Of these, heart disease (61.36 %) and vascular disease (34.09 %) were the primary contributors to CVD mortality in DMCA patients (Fig. 1G). In the subgroup analyses, both cancer patients with pre-existing diabetes and cancer patients diagnosed with diabetes during follow-up exhibited significantly higher risks of all-cause mortality and cardiovascular mortality among cancer patients with diabetes undergoing chemotherapy (DMCA patients) (Figure S1 and Figure S2 both show P < 0.001).

Fig. 1.

Fig. 1

Prevalence, prognosis, and cardiovascular damage in cancer patients with diabetes receiving chemotherapy. (A) Proportion of cancer patients with diabetes (DMCA) undergoing chemotherapy from the UK Biobank. (B) Kaplan-Meier (KM) survival curve comparing overall survival (OS) between DMCA and cancer patients without diabetes (CA). (C) Kaplan-Meier (KM) survival curve comparing cardiovascular disease (CVD) incidence between DMCA and CA. (D) Cox regression analysis for OS comparing DMCA with CA. The crude model was unadjusted; Model 1 was adjusted for age, sex, and race; Model 2 was adjusted for Model 1 plus BMI, smoking status, drinking status, income, education, hypertension, and hyperlipidemia. (E) Cox regression analysis for CVD comparing DMCA with CA. The crude model was unadjusted; Model 1 was adjusted for age, sex, and race; Model 2 was adjusted for Model 1 plus BMI, smoking status, drinking status, income, education, hypertension, and hyperlipidemia. (F) Mortality from non-cancer causes among cancer patients with diabetes undergoing chemotherapy. (G) Mortality from CVD causes among cancer patients with diabetes undergoing chemotherapy.

In summary, this prospective cohort study clearly demonstrates that heart and vascular diseases are significant health challenges faced by patients with diabetes and those undergoing cancer chemotherapy. Therefore, we further focused on drug screening targeting the treatment of heart and vascular injuries.

2.2. Design of silicified curcumin (Si/Cur) microspheres based on key targets of heart and vascular injury

We conducted a network pharmacological analysis to screen compounds targeting key factors in heart and vascular injury. To visually illustrate the drug-target interactions, we created a drug-target network diagram (Fig. 2A). This diagram comprises 366 nodes, each representing a disease target, with the connecting lines indicating the association between drugs and their respective targets. A Venn diagram was used to identify the common drugs for cardiac and vascular injuries, resulting in 909 drugs that treat both conditions. Among these, 30 drugs with at least 25 effective target interactions were retained. From these 30 drugs, four TCMs were screened, including resveratrol, genistein, quercetin, and curcumin (Table S2). The total interaction score for all targets indicated that curcumin ranked second in terms of cardiac injury, and it also showed efficacy in vascular injury (Table S2). To select the most suitable candidate drug from the four compounds, we first evaluated two important physicochemical parameters using the Traditional Chinese Medicine Systems Pharmacology Database and Analysis Platform (TCMSP) [35]: the partition coefficient (AlogP) and drug-likeness (DL) (Table S3). The AlogP value reflects the balance between a compound's lipophilicity and hydrophilicity, which determines its absorption and distribution in the body. It is generally believed that an AlogP value between 2 and 5 is ideal, as compounds within this range exhibit good absorption characteristics. Among the four compounds, we found that quercetin had an AlogP value of only 1.5, which is below this standard. Such low value suggests that the compound's bioavailability may be insufficient, thereby limiting its efficacy in the body. Therefore, quercetin was excluded. Next, we focused on assessing the DL value, which measures a compound's potential as a drug based on its molecular properties. Compounds with a DL value greater than 0.18 are generally considered to have good prospects for drug development. Although resveratrol performed well in the target interaction scoring, its DL value was only 0.11, significantly lower than the standard, and thus it was also eliminated. Ultimately, two candidate compounds remained: curcumin and genistein. Both compounds met the screening criteria for AlogP and DL values, but further comparison showed that curcumin performed more excellently overall. Its AlogP (3.36) and DL (0.41) values were within the ideal range, and its total scores for cardiac injury (3.7241) and vascular injury (0.0961) targets were also better than those of genistein (Table S2). Curcumin (Cur), a well-established natural cardioprotective agent, has demonstrated a strong antifibrotic effect in broad investigations. Therefore, we selected Cur for further research.

Fig. 2.

Fig. 2

Employing a data mining approach (data on therapeutic drug targets along with vascular and heart injuries) to guide the design of silicified curcumin (Si/Cur). (A) A Venn diagram depicting the overlap between potential drug candidates for heart injury and vasculitis. (B) A drug-target network diagram. (C) Photographic images of curcumin in Si ion solutions at various concentrations. (D) UV absorption peaks and solubility profiles of curcumin in Si ion solutions across different concentrations. (E) Scanning electron microscopy, transmission electron microscopy, and elemental distribution maps of the Si/Cur microsphere. (F) Release of Si ions and Cur from the Si/Cur microspheres. (G–I) Scavenging effects of Si/Cur on ABTS, H2O2, and •O2.

However, Cur has limitations including low solubility and bioavailability of Cur, though its solubility improves in alkaline solutions. Building on previous findings from our research group, we know that Si ion solutions can generate an alkaline microenvironment with notable biological activity [36]. Thus, we hypothesized that Si ion-rich solutions could enhance Cur solubility. To test this, we mixed an excess of curcumin with Si ion solutions at varying concentrations, creating what we refer to as Si/Cur solutions. Notably, visual inspection revealed that as the concentration of Si ions in the solution increased, the amount of solubilized curcumin also rose, resulting in a deeper red hue (Fig. 2B). This trend was further confirmed by UV spectrophotometry, where higher Si ion concentrations corresponded to increased absorbance at 425 nm, indicating higher concentrations of solubilized curcumin (Fig. 2C and D). Next, we fabricated the Si/Cur into composite microsphere biomaterials (Si/Cur microspheres). Scanning electron microscopy (SEM) revealed that these Si/Cur microspheres had a particle size of approximately 50 nm and were rich in calcium and silicon elements (Fig. 2E). The SEM images reveal that the particle size of the Si/Cur microspheres is highly uniform. This is attributed to the preparation method, where Si microspheres are first synthesized via the sol-gel method and then loaded with Cur. As a result, the particle size of the Si/Cur microspheres remains consistent and is not altered by changes in the concentration of Cur or Si microspheres. The drug release profile demonstrated that curcumin was effectively released over 14 days as Si ions were released, meeting the requirement for sustained delivery (Fig. 2F). Furthermore, the Si/Cur released from the microspheres did not diminish the inherent free radical scavenging ability of curcumin. Remarkably, we found that the scavenging activity of Si/Cur against ABTS, H2O2, and •O2 was comparable to that of curcumin (Fig. 2G–I).

In summary, Si ion-rich solutions can effectively enhance the solubility of curcumin, potentially improving its bioavailability. Simultaneously, this Si/Cur can be delivered in the form of microspheres while maintaining excellent free radical scavenging ability.

2.3. Therapeutic effects of Si/Cur Microspheres on diabetic vascular remodeling and doxorubicin-induced cardiotoxicity

To validate the therapeutic effectiveness of Si/Cur Microspheres, we established a diabetic mouse model treated with DOX to simultaneously simulate vascular remodeling and DOX-induced cardiomyopathy. We then assessed the therapeutic impacts of Si/Cur microspheres on both types of injury. For diabetic vascular remodeling, we observed changes in vascular structure, collagen fibers, and elastic fibers using HE staining, Masson staining, and Victoria blue staining (Fig. 3A). The results showed that Si/Cur microspheres could significantly inhibit the increase in vascular wall thickness (intima-media) in a high-glucose environment, outperforming the Si group alone (Fig. 3B). In diabetic mice, the collagen fibers in the vascular wall were notably increased, with cross-linking between fibers and visible block-like fibrotic areas between elastic plates. However, these fibrotic phenomena were markedly reduced under the intervention of Si/Cur microspheres (Fig. 3C). Compared to the control group, the decrease in vascular elastic fibers caused by high-glucose was also improved with Si/Cur treatment (Fig. 3D). Immunohistochemical observations further revealed that Si/Cur significantly inhibited the expression of inflammatory factors ICAM-1 and VCAM-1 on the vascular intima in a high-glucose environment (Fig. 3E–G).

Fig. 3.

Fig. 3

Therapeutic effects of Si/Cur microspheres on the mice with mixed diseases of diabetic vascular remodeling and doxorubicin-induced cardiomyopathy. (A) Histological analysis (H&E, Masson and Victoria staining) of aortic vessels from the mice treated with Si/Cur microspheres. (B–D) Quantitative analysis of aortic intima-media thickness (B), fibrotic area (C), and elastic fiber area (D) in the aortic wall. (E) immunohistochemical analysis (ICAM-1 and VCAM-1) of aortic vessels from the mice treated with Si/Cur microspheres. (F, G) Quantitative analysis of ICAM-1 protein expression (F) and VCAM-1 protein expression (G) in the aortic wall. (H) Representative M-mode echocardiographic images of the left ventricle following Si/Cur microsphere treatment. (I) Dynamic changes in ejection fraction and fractional shortening after Si/Cur microsphere treatment. (J) Masson's trichrome images of heart areas post Si/Cur microsphere treatment. (K) Statistical analysis of the percentage of Masson's trichrome-positive area in the heart after Si/Cur microsphere treatment. (L) Heart weight-to-tibia length ratio of mice after Si/Cur microsphere treatment. (M) TUNEL staining of myocardial regions after Si/Cur microsphere treatment. (N) Quantitative analysis of TUNEL-positive cell counts. Groups: Sham: normal mice. Control: mice with mixed diseases of diabetic vascular remodeling and doxorubicin-induced cardiomyopathy. Si: mice with mixed diseases treated with Si microspheres. Si/Cur: mice with mixed diseases treated with Si/Cur microspheres. (&P < 0.05 vs Control, ∗P < 0.05 vs Si, n = 5).

For the doxorubicin-induced cardiotoxicity model, we evaluated left ventricular systolic function using echocardiography. The results indicated that Si/Cur microspheres improved the decrease in ejection fraction (EF) caused by DOX, outperforming the Si group. Additionally, Si/Cur microspheres improved fractional shortening (FS), while the Si group had no significant therapeutic effect on FS (Fig. 3H–I). To assess the impact of Si/Cur on myocardial fibrosis, we used Masson staining to analyze rat heart tissue sections (Fig. 3J). The results showed significant heart fibrosis in the DOX group, while both the Si group and the Si/Cur group inhibited the increase in the fibrotic area under DOX, with the Si/Cur group showing a more pronounced inhibitory effect (Fig. 3K). By measuring the heart weight to tibia length ratio (HW/TL), we found that both the Si/Cur group and the Si group preserved heart weight compared to the control group, with the Si/Cur group exhibiting a stronger improvement effect (Fig. 3L). TUNEL staining results further demonstrated that the Si/Cur group significantly inhibited DOX-induced cardiomyocyte apoptosis, outperforming the Si group (Fig. 3M–N).

Lastly, we evaluated the biosafety of Si/Cur microspheres. It is crucial to note that Si/Cur did not cause any damage to organs such as the liver, spleen, lungs, and kidneys, indicating good biological safety (Fig. S3). Following Si/Cur intervention, we used the ELISA method to measure liver and kidney function markers in mice (Fig. S4). At the experiment's initiation, there were no significant differences in liver and kidney function between Control and Sham mice. As the disease progressed, no notable abnormalities in liver and kidney function were detected in the Control group. Importantly, compared to other groups, the Control group treated with Si/Cur did not show any significant changes in liver and kidney function. These results further confirm the biosafety of Si/Cur microspheres and support their potential for clinical translation.

2.4. Synergistic effect of Si/Cur solution in the treatment of diabetic vascular remodeling

To ascertain that the primary therapeutic agent within Si/Cur microspheres, responsible for eliciting the core therapeutic effect, is indeed the synergistic effect of Si and Cur in the soluble silicified curcumin (Si/Cur), we further evaluated the therapeutic potential of Si/Cur in db/db diabetic mice, comparing it with Cur alone (Cur) and Si alone. Our experimental results indicated that the diabetic (Control) mice exhibited increased body size and significant obesity (Fig. 4A). Over the 8-week experimental period, Control group consistently maintained higher body weight and blood glucose levels compared to non-diabetic (Sham) mice (Fig. 4B–C). Notably, the mice treated with Si/Cur demonstrated a slight decrease in blood glucose levels at week 6 and a significant reduction by week 8, in contrast to the control, Cur, and Si groups (Fig. 4C). Analysis of inflammatory factors revealed that at the study's onset, Control group had elevated levels of IL-1β, IL-6, and TNF-α in their blood compared to Sham mice. These inflammatory markers continued to increase over time in Control group. Throughout the intervention period, other groups also exhibited significantly elevated levels of inflammatory factors compared to the Sham group. However, treatment with Si/Cur or Si significantly reduced these inflammatory markers in Control group, with Si/Cur demonstrating a more pronounced effect (Fig. 4D). Histological examination of the thoracic aorta using H&E staining revealed no significant difference in wall thickness between Sham and Control group at the start of the experiment. During the study, the walls of Control group showed significant thickening, with proliferation protruding into the lumen and the presence of vacuolated foam cells. Both Si/Cur and Si treatments effectively inhibited this wall thickening in diabetic mice, with Si/Cur demonstrating greater efficacy than Si alone. Curcumin (Cur) did not significantly impact diabetes-induced wall thickening (Fig. 4E). Further analysis utilizing Masson staining and Victoria blue staining techniques revealed that Si/Cur significantly reduced fibrosis to a greater extent, whereas Si ion exhibited a more pronounced reduction in elastic fibers within the aortic walls of diabetic mice (Fig. 4E). Quantitative statistical analysis corroborated these findings, indicating that Si/Cur exerted the most marked effect on diminishing wall thickening and fibrosis. Additionally, both Si ion and Si/Cur demonstrated an impact on elastic fiber degradation in the Control group (Fig. 4F–H).

Fig. 4.

Fig. 4

Impact of Si/Cur on vascular remodeling in diabetic mice. (A) Photos of diabetic (Control) and non-diabetic (Sham) mice. (B) Body weight variations in diabetic mice upon Si/Cur treatment. (C) Fasting blood glucose trends in mice. (D) The expression of inflammatory factors (IL-6, IL-1β, and TNF-α) in serum after Si/Cur treatment. (E) Histological analysis (H&E, Masson, and Victoria Blue staining) of aortic vessels in diabetic mice treated with Si/Cur to assess intima-media thickness, and collagen/elastic fiber content. (F–H) Quantitative analysis of aortic intima-media thickness (F), collagen fiber area (G) and elastic fiber area (H) in diabetic mice. Groups: Sham: non-diabetic mice. Control: diabetic mice. Cur: diabetic mice treated with Cur. Si/Cur: diabetic mice treated with Si/Cur. Si: diabetic mice treated with Si. (&P < 0.05 vs Sham Day 0, @P < 0.05 vs Control Day 0, $ P < 0.05 vs Sham, ∗P < 0.05 vs Control, #P < 0.05 vs Si and Cur, n = 5.

ICAM-1 and VCAM-1 adhesion molecules are essential for facilitating the infiltration of inflammatory cells into vascular walls. To explore this mechanism further, we performed immunohistochemical staining to evaluate the expression of ICAM-1 and VCAM-1 in the vascular walls. Initially, there was no significant difference in the expression levels of ICAM-1 and VCAM-1 between normal blood glucose (Sham) and high blood glucose (Control) mice. However, over time, we observed a significant increase in the expression of both ICAM-1 and VCAM-1 in the vascular walls of Control group compared to Sham mice. The application of Si/Cur and Si significantly inhibited these adhesion molecules in diabetic mice, with Si/Cur demonstrating a more pronounced inhibitory effect. In contrast, Cur alone did not significantly reduce the upregulation of ICAM-1 and VCAM-1 induced by diabetes (Fig. 5A–S5A, and S5B). To accurately quantify the expression of ICAM-1 and VCAM-1 proteins in the vessels of diabetic mice, a Western blot immunoassay was performed. Before intervention, ICAM-1 expression was already elevated in Control group, while the VCAM-1 protein level showed a slight increase that was not statistically significant compared to Sham mice (Fig. 5B–E). As vascular remodeling progressed, the protein levels of ICAM-1 and VCAM-1 continued to significantly increase in Control group. In contrast, both the Si and Si/Cur groups demonstrated notable inhibitory effects on the expression of ICAM-1 and VCAM-1, with Si/Cur showing a stronger effect. Given the critical role of ICAM-1 and VCAM-1 in the infiltration of inflammatory cells into vascular walls, we further analyzed the immunofluorescence results of CD68-positive cells in the thoracic aorta of the mice. At the beginning of the experiment, there was no significant difference in the proportion of CD68-positive cells between Sham and Control group. However, over time, the proportion of CD68-positive cells in the vascular walls of Control group significantly increased compared to the Sham group, and this elevated proportion persisted even after the intervention phase. Importantly, treatment with Si/Cur and Si resulted in a significant decrease in the proportion of CD68-positive cells in the vascular walls of diabetic mice, with Si/Cur demonstrating a more pronounced reduction (Fig. 5F and S5C).

Fig. 5.

Fig. 5

Si/Cur regulates adhesion molecules in diabetic vascular walls to inhibit the infiltration of inflammatory factors in vascular walls (A) Immunohistochemical staining reveals the effects of Si/Cur on ICAM-1 and VCAM-1 protein expression in the aortic wall of diabetic mice. (B, C) Western Blot analysis and quantitation of ICAM-1 protein levels in the aorta of diabetic mice treated with Si/Cur. (D, E) Western Blot analysis and quantitation of VCAM-1 protein levels in the aorta of the diabetic mice treated with Si/Cur. (F) Impact of Si/Cur on CD68-positive cell infiltration in the aortic vessels of diabetic mice. Groups: Sham: non-diabetic mice. Control: diabetic mice. Cur: diabetic mice treated with Cur. Si/Cur: diabetic mice treated with Si/Cur. Si: diabetic mice treated with Si. (&P < 0.05 vs Sham Day 0, @P < 0.05 vs Control Day 0, $ P < 0.05 vs Sham, ∗P < 0.05 vs Control, #P < 0.05 vs Si and Cur, n = 5).

2.5. Synergistic effect of Si/Cur solution on doxorubicin-induced cardiotoxicity

In the doxorubicin (DOX)-induced cardiotoxicity model, we initially monitored changes in mouse body weight. The results showed that, while the mice treated with DOX experienced a significant decrease in body weight, the Cur group, Si group, and the Si/Cur combination therapy group all exhibited weight recovery, with the most pronounced improvement seen in the Si/Cur group (Fig. 6A). Next, we assessed cardiac function in mice using echocardiography. The results revealed that left ventricular systolic function was markedly reduced in the mice treated with DOX, whereas in the Si/Cur combination therapy group, left ventricular systolic function was significantly improved. Changes in the Cur group and Si group, however, did not reach statistical significance (Fig. 6B–C). By measuring the ratio of heart weight to tibia length (HW/TL), we found that heart weight was better preserved in the Si/Cur combination therapy group compared to the control group (Fig. 6D). In contrast, neither the Cur group nor the Si group demonstrated significant improvement in DOX-induced cardiac atrophy. To further evaluate the impact of Si/Cur combination therapy on myocardial fibrosis, we performed Masson's trichrome staining on rat heart tissue sections (Fig. 6E). The results demonstrated that the Si/Cur combination therapy group significantly reduced the increase in myocardial fibrosis area induced by DOX (Fig. 6E–F). TUNEL staining further revealed that this therapy group exhibited the most pronounced effect in inhibiting DOX-mediated cardiomyocyte apoptosis (Fig. 6G–H). Additionally, compared to the control group, the Si/Cur combination therapy group significantly lowered the levels of cardiac injury markers, including CK-MB, cTnT, and LDH in the plasma (Fig. 6I–K).

Fig. 6.

Fig. 6

Impact of Si/Cur on mitigating myocardial injury in mice with doxorubicin-induced cardiomyopathy. (A) Variations in body weight of the mice with doxorubicin-induced cardiomyopathy upon Si/Cur treatment. (B) Representative M-mode echocardiographic images of the left ventricle following Si/Cur treatment. (C) Dynamic changes in ejection fraction and fractional shortening after Si/Cur treatment. (D) Heart weight-to-tibia length ratio of mice after Si/Cur treatment. (E) Masson's trichrome images of heart areas post Si/Cur treatment. (F) Statistical analysis of the percentage of Masson's trichrome-positive area in the heart after Si/Cur treatment. (G) TUNEL staining of myocardial regions after Si/Cur treatment. (H) Quantitative analysis of TUNEL-positive cell counts. (I–K) Expression levels of myocardial injury serum markers CK-MB, cTnT, and LDH after Si/Cur treatment. Groups: Sham: normal mice. Control: mice with doxorubicin-induced cardiomyopathy. Si: mice with doxorubicin-induced cardiomyopathy treated with Si. Cur: mice with doxorubicin-induced cardiomyopathy treated with Cur. Si/Cur: mice with doxorubicin-induced cardiomyopathy treated with Si/Cur. (&P < 0.05 vs Control, ∗P < 0.05 vs Si, #P < 0.05 vs Cur).

2.6. Protection of human umbilical vein endothelial cells (HUVECs) from high-glucose-induced damage by Si/Cur via the Nrf2/HO-1 signaling pathway

To delve into the mechanisms underlying the effects of Si/Cur on diabetic vascular remodeling, we simulated a high-glucose environment to induce damage in human umbilical vein endothelial cells (HUVECs) by administering glucose. Initial experiments revealed that, compared to 33 mmol/L glucose, 50 mmol/L glucose significantly reduced cell viability. Therefore, we chose 50 mmol/L as the glucose concentration to establish a cell model of high-glucose-induced damage (Fig. S6) before treating the affected HUVECs with Si/Cur.

The experimental results demonstrated that, under high-glucose conditions, the protein and RNA expression of adhesion molecules ICAM-1 and VCAM-1 in HUVECs significantly increased compared to the normal group. Treatment with Si, Cur, and Si/Cur notably inhibited the expression of these adhesion molecules, with Si/Cur demonstrating the most pronounced effect, surpassing those of Si and Cur alone (Fig. 7A–F). The elevated levels of ICAM-1 and VCAM-1 triggered by the high-glucose environment further induced oxidative stress responses in HUVECs (Fig. 7G), resulting in a substantial accumulation of reactive oxygen species (ROS) within the cells. Importantly, Si, Cur, and Si/Cur effectively scavenged these ROS, with Si/Cur demonstrating the most potent scavenging ability (Fig. 7H).

Fig. 7.

Fig. 7

Si/Cur solution protects HUVECs from high-glucose damage. (A, B) Western Blot analysis and quantitation of ICAM-1 protein levels in HUVECs treated with Si/Cur under high-glucose conditions. (C, D) Western Blot analysis and quantitation of VCAM-1 protein levels in HUVECs treated with Si/Cur under high-glucose conditions. (E, F) RNA expression of ICAM-1 and VCAM-1 in HUVECs treated with Si/Cur under high-glucose conditions. (G, H) Assessment of oxygen radical production in HUVECs treated with Si/Cur under high-glucose conditions. (I, J) Western Blot analysis and quantitation of HO-1 protein levels in HUVECs treated with Si/Cur under high-glucose conditions. (K, L) Western Blot analysis and quantitation of Nrf2 protein levels in HUVECs treated with Si/Cur under high-glucose conditions. (M) Cellular distribution of Nrf2 protein in the cytoplasm and nucleus of HUVECs treated with Si/Cur under high-glucose conditions. Groups: Blank: HUVECs cultured under normal culture medium. Control: HUVECs cultured under high-glucose culture medium. Cur: HUVECs cultured under high-glucose culture medium and treated with Cur Si/Cur: HUVECs cultured under high-glucose culture medium and treated with Si/Cur. Si: HUVECs cultured under high-glucose culture medium and treated with Si. ($ P < 0.05 vs Blank, ∗P < 0.05 vs Control, #P < 0.05 vs Si and Cur, n = 5).

Furthermore, the high-glucose environment also led to increased expression of inflammatory factors, including IL-6, IL-1β, and TNFα, in HUVECs. Treatment with Si, Cur, and Si/Cur significantly reduced the levels of these inflammatory factors, with Si/Cur showing the most pronounced reduction effect (Fig. S7). To further investigate the adhesion between endothelial cells and inflammatory cells under high-glucose conditions, as well as the impact of Si/Cur on this adhesion, we conducted an in vitro adhesion experiment using monocytes (THP-1). Fluorescence microscopy observations revealed that the high-glucose environment significantly enhanced the adhesion of THP-1 cells to HUVECs. However, treatment with Si, Cur, and Si/Cur, led to a significant decrease in cell adhesion, with Si/Cur exhibiting the most pronounced effect (Fig. S8).

HO-1 is a key protease involved in antioxidant stress and inflammation inhibition, while Nrf2 is a crucial regulator of HO-1 protein expression. Furthermore, the distinctive structure within the Cur molecule possesses the ability to bind to essential residues of the Keap1 protein, initiating a cascade of reactions that ultimately culminate in the activation of Nrf2 and the transcription of antioxidant genes [37,38]. Consequently, we postulate that Si/Cur may exert therapeutic effects via the Nrf2/HO-1 signaling pathway.

To elucidate whether Si, Cur, and Si/Cur exert their antioxidant and anti-inflammatory effects through the HO-1 enzyme, we performed a semi-quantitative analysis of HO-1 protein expression via protein electrophoresis (Fig. 7I–J). Under high-glucose stimulation, HO-1 expression compensatorily increased compared to the normal group. Compared to the control, treatment with Si, Cur, and Si/Cur further enhanced HO-1 expression in the cells, with Si/Cur demonstrating the most significant upregulation. Fig. 7K–L indicate that Si/Cur can also significantly upregulate the expression of the transcription factor Nrf2. Nrf2, a member of the transcription factor family, primarily binds to the Keap1 protein under physiological conditions, remaining in the cytoplasm and unable to enter the nucleus to regulate proteins like HO-1. However, under stress conditions, the Keap1 protein degrades, enabling the Nrf2 protein to migrate into the nucleus and exert its functions. To gain a deeper understanding of the regulatory mechanisms of Si/Cur on Nrf2, we observed the distribution of Nrf2 inside and outside the nucleus using immunofluorescence. Fig. 7M and S9 show that under high-glucose conditions, the fluorescence of Nrf2 in the nucleus increased slightly, which is greater in the Si/Cur group than in the control. By analyzing the nuclear and cytoplasmic distribution of Nrf2, we found that the proportion of Nrf2 protein in the nucleus significantly increased in both the control and Si/Cur groups, with Si/Cur further enhancing the nuclear translocation of Nrf2.

2.7. Protection of smooth muscle cells (SMCs) from high-glucose induced damage by Si/Cur through the Nrf2/HO-1 signaling pathway

Exposure to a high-glucose environment can impair the contractile function of vascular smooth muscle and trigger a phenotypic shift from contractility to synthesis. This phenotypic modulation of SMCs plays a central role in vascular remodeling, atherosclerotic plaque formation, and vascular stenosis. Therefore, we investigated the protective effects of Si/Cur on SMCs under high-glucose (control) conditions.

Enhanced SMCs migration is a notable hallmark of phenotypic transformation under high-glucose exposure. Scratch assay results revealed that SMCs in a high-glucose (control) environment exhibited significantly enhanced migratory ability compared to control SMCs. However, compared to the Control, treatment with Cur, Si/Cur, or Si alone effectively attenuated this migration, with no significant differences in migration inhibitory effects among the three treatment methods (Fig. 8A–B). Phenotypic modulation of SMCs is a critical pathological change in diabetic vascular remodeling, characterized by a decrease in α-SMA expression, which is a key indicator of the shift from contractility to synthesis. We assessed the effects of Cur, Si/Cur, and Si on SMC phenotypic modulation through Western blot analysis. Under high-glucose (Control) conditions, α-SMA expression was significantly reduced compared to normal-glucose (Blank) conditions. Compared to cells in a high-glucose environment, all three treatment methods restored α-SMA levels, with Si/Cur showing a more pronounced effect in counteracting high-glucose-induced changes in protein expression (Fig. 8C–D). SMCs not only respond to inflammatory signals from other cells but also secrete inflammatory mediators such as IL-1β and IL-6 under stress, actively participating in vascular remodeling. Our data showed that high-glucose significantly upregulated the release of these inflammatory factors. However, treatment with Cur, Si/Cur, and Si effectively attenuated this high-glucose-induced inflammatory response, with Si/Cur showing the most significant therapeutic effect (Fig. 8E and F).

Fig. 8.

Fig. 8

Si/Cur protects SMCs from high-glucose damage. (A) Migration of SMCs under high-glucose conditions with Si/Cur treatment (B) Quantitative statistical analysis of SMCs migration rate. (C, D) Western Blot analysis and quantitation of α-SMA protein levels in SMCs treated with Si/Cur under high-glucose conditions. (E, F) Impact of Si/Cur on inflammatory factor production in SMCs under high-glucose conditions. (G, H) Western Blot analysis and quantitation of HO-1 protein levels in SMCs treated with Si/Cur under high-glucose conditions. (I, J) Western Blot analysis and quantitation of Nrf2 protein levels in SMCs treated with Si/Cur under high-glucose conditions. Groups: Blank: SMCs cultured under normal culture medium. Control: SMCs cultured under high-glucose culture medium. Cur: SMCs cultured under high-glucose culture medium and treated with Cur. Si/Cur: SMCs cultured under high-glucose culture medium and treated with Si/Cur. Si: SMCs cultured under high-glucose culture medium and treated with Si. ($ P < 0.05 vs Blank, ∗P < 0.05 vs Control, #P < 0.05 vs Si and Cur, n = 5).

To further explore the mechanism of action of Si/Cur, we investigated the expression of HO-1 and its upstream regulator Nrf2. The results indicated that both proteins were compensatorily upregulated under high-glucose (Control) conditions compared to normal-glucose (Blank) conditions. Treatment with Cur, Si/Cur, and Si further enhanced the expression of these proteins, with Si/Cur showing the most significant therapeutic effect (Fig. 8G, H, 8I and 8J).

2.8. Si/Cur protects doxorubicin-induced cardiotoxicity via the Nrf2/HO-1 signaling pathway

After administration of DOX, a substantial quantity of ROS is generated within NRCMs, ultimately resulting in their apoptosis. This process leads to a significant reduction in cardiomyocyte viability and a notable increase in apoptosis. Nevertheless, intervention with Si/Cur effectively prevents the elevation of ROS levels, mitigates the decline in cell viability, and suppresses apoptosis. In contrast, neither the Si group nor the Cur group alone significantly affected the DOX-induced decrease in cell viability or increase in apoptosis (Fig. 9A–D). To further investigate the mechanism, we analyzed the expression of HO-1 protein and Nrf2 using Western Blot. The results showed that after DOX treatment, the expression of HO-1 increased compensatorily. Compared with the control group, Si/Cur further increased the expression of HO-1 in cells, and the expression of Nrf-2 also increased. However, neither the Cur group nor the Si group alone showed significant differences in the regulation of HO-1 compared with the control group (Fig. 9E–H). Furthermore, Fig. 9I–J illustrate that under DOX induction, Nrf2 fluorescence in the NRCMs nucleus increased slightly. However, the fluorescence enhancement in the Si/Cur group surpassed that of the control. By examining the nuclear and cytoplasmic distribution of Nrf2, we discovered that the proportion of Nrf2 protein in the nucleus significantly increased in both the control and Si/Cur groups, with Si/Cur further enhancing the nuclear translocation of Nrf2.

Fig. 9.

Fig. 9

Si/Cur mitigates DOX-induced cardiomyocytes (NRCMs) injury and enhances cell viability. (A, B) Si/Cur inhibits the generation of reactive oxygen species (ROS) in NRCMs induced by DOX. (C) NRCMs viability following DOX-induced injury and subsequent Si/Cur treatment. (D) Apoptosis in DOX-induced NRCMs after Si/Cur treatment. (E, F) Western blot analysis showing the expression levels of HO-1 and Nrf2 in DOX-injured NRCMs after Si/Cur treatment. (G, H) Quantitative analysis of HO-1 and Nrf2 expression levels in DOX-injured NRCMs following Si/Cur treatment. (I, J) Cellular distribution of Nrf2 protein in the cytoplasm and nucleus of NRCMs treated with Si/Cur. Quantitative analysis of Nrf2 protein distribution in the cytoplasm and nucleus of NRCMs. Groups: Blank: cardiomyocyte cultured under normal culture medium. Control: cardiomyocyte cultured under DOX culture medium. Cur: cardiomyocyte cultured under DOX culture medium and treated with Cur. Si/Cur: cardiomyocyte cultured under DOX culture medium and treated with Si/Cur. Si: cardiomyocyte cultured under DOX culture medium and treated with Si. (& P < 0.05 vs Control, ∗P < 0.05 vs Si, #P < 0.05 vs Cur, n = 5).

3. Discussion

Cancer patients with concomitant diabetes face dual challenges after chemotherapy: the cardiotoxic effects of chemotherapeutic agents [16,17] and vascular remodeling associated with diabetes itself [5,6], posing threats to both the heart and vasculature. Unfortunately, this subpopulation of patients with comorbid conditions has not received sufficient clinical attention. This oversight is primarily due to the siloed nature of clinical focus, where oncologists often prioritize cancer treatment and prognosis [39,40], while endocrinologists concentrate on diabetes management [41]. Such disciplinary differences may hinder the delivery of comprehensive care to these patients with dual pathologies. This issue is further exacerbated by the limited theoretical evidence elucidating the specific cardiovascular risks faced by diabetic cancer patients undergoing chemotherapy. As a result, this complex subpopulation is more susceptible to being overlooked. They are often treated as typical oncology patients, which may worsen their condition and contribute to increased mortality rates. To address this concern, our study proposes a prospective cohort investigation comparing diabetic cancer patients receiving chemotherapy with their non-diabetic counterpart. The aim is to thoroughly analyze the prevalence, prognosis, and cardiovascular damage in diabetic cancer patients undergoing chemotherapy. Through such analysis, we can quantify the number of patients with these comorbid conditions and identify the leading causes of mortality, with the hope of drawing widespread clinical attention to these issues. Notably, between 2006 and 2022, the proportion of diabetic patients among cancer patients receiving chemotherapy has significantly risen from 2.4 % to 10.8 %. This trend highlights the growing number of patients with these comorbid conditions, underscoring the urgency of addressing this population's unique needs. Even more alarming is our finding that the primary causes of mortality for diabetic cancer patients undergoing chemotherapy are not cancer itself, but heart disease (accounting for 61.36 %) and vascular disorders (accounting for 34.09 %). This revelation underscores the pressing need for designing new treatment strategies to address the cardiac and vascular issues faced by the growing number of diabetic cancer patients undergoing chemotherapy, which has become a significant clinical concern requiring immediate attention.

Through an extensive analysis of the pathophysiological mechanisms affecting cancer patients with diabetes who receive chemotherapy, we have observed that under specific conditions such as hyperglycemia, endothelial cells in the human body are activated, leading to abnormal overexpression of adhesion molecules on their surface, including ICAM-1 and VCAM-1, along with a substantial release of inflammatory factors. These inflammatory factors not only induce the adhesion and infiltration of inflammatory cells from the circulation into the vessel wall but also trigger inflammatory responses in other cells within the vessel wall [[13], [14], [15]]. Further complicating this scenario, these inflammatory factors synergize with chemotherapeutic agents, directly attacking NRCMs and inducing their extensive apoptosis, ultimately resulting in cardiac dysfunction [16,17]. On the other hand, they exacerbate the release of additional inflammatory factors from other cells within the vessel wall, creating a vicious inflammatory cycle that is difficult to break [[42], [43], [44], [45]]. Given the intricate pathological conditions of this comorbid disease, we believe that relying solely on anti-inflammatory or cardioprotective drugs may be inadequate for effective management. To design a treatment strategy capable of disrupting this vicious cycle, our study conducted thorough data mining using the DisGeNET, STRING, and DGIdb databases and identified curcumin (Cur) as a potential therapeutic agent through network pharmacological analysis. We further propose utilizing silicon-based biomaterials to construct a Si ion alkaline microenvironment, maximizing the bioavailability of Cur [36]. The research findings indicate that the remarkable antioxidant and anti-inflammatory properties of Cur [[20], [21], [22]], combined with the significant efficacy of silicon-based biomaterials in improving vascular-related diseases [27], indeed offer an effective strategy for addressing the cardiac and vascular issues associated with this comorbid disease.

High-glucose levels are a crucial factor triggering vascular remodeling in diabetes, particularly during the initial stages, where inflammatory damage to endothelial cells plays a pivotal role. This inflammatory response is manifested by a significant increase in the expression and release of adhesion molecules ICAM-1 and VCAM-1, along with a substantial elevation in the release of key inflammatory cytokines such as IL-1β, IL-6, and TNFα [[46], [47], [48]]. In our experiments, treatment with Si, Cur, and Si/Cur led to a notable reduction in the production of these two adhesion molecules and the three inflammatory cytokines. Additionally, by measuring ROS, we found that the anti-inflammatory effects of these treatments might be related to their inhibition of oxidative stress induced by high-glucose levels. Monocyte adhesion assays clearly demonstrated the significant efficacy of the Si/Cur combination in ameliorating endothelial cell inflammation and mitigating inflammatory cell infiltration in the vessel wall. The anti-inflammatory effects exhibited by Cur in the experiments were consistent with other research findings, and the addition of Si further enhanced these effects. In our initial exploration of the anti-inflammatory mechanisms of Si/Cur, we discovered that its pronounced anti-inflammatory action was closely associated with the upregulation of Nrf2 expression, which subsequently enhanced cellular HO-1 expression. The Nrf2/HO-1 signaling pathway is currently recognized as an important pathway involved in cellular anti-inflammatory and antioxidant regulation. Additionally, smooth muscle cells are a core component of muscular arteries. Distinct from endothelial cells, vascular endothelium may experience pathological changes such as damage, apoptosis, or necrosis when confronted with challenges like hyperglycemia, inflammation, and oxidative stress. Smooth muscle cells, however, adopted a different coping strategy: they may dedifferentiate, enhance proliferation and migration capabilities, reduce contractility, and augment synthetic and secretory functions [[49], [50], [51]]. These changes can ultimately lead to vessel wall thickening and luminal stenosis [[52], [53], [54]]. Through scratch assays, we observed that a high-glucose environment accelerates the migration of vascular smooth muscle cells. Yet interestingly, our study is the first to reveal that, under high-glucose conditions, Si extract did not promote the migration of vascular smooth muscle cells; rather, it appeared to decelerate it. Meanwhile, our experimental results indicated that Si/Cur, Si, and Cur could all effectively inhibit cell migration, although the differences between the groups did not reach statistical significance. Furthermore, Si/Cur, Si, and Cur could also suppress the production of inflammatory cytokines IL-1β and IL-6 induced by high-glucose levels and prevent the downregulation of α-SMA protein expression, with Si/Cur demonstrating the most prominent effects. To gain a deeper understanding of its mechanisms, we conducted further research and found that for smooth muscle cells, the significant effects of Si/Cur on anti-inflammation, inhibiting cell migration, and improving contractile function during phenotypic transformation might also be related to the Nrf2/HO-1 signaling pathway. This discovery enhances our understanding of the mechanisms of Si/Cur in treating diabetic vascular remodeling and provides new directions for future research on diabetic vascular complications.

Numerous studies have clearly indicated that the chemotherapeutic drug doxorubicin (DOX) directly damages NRCMs, thereby triggering significant myocardial toxicity [55,56]. However, when administered in the context of diabetes, the cardiac toxicity of DOX may be further exacerbated. This occurs because diabetes, a metabolic syndrome characterized by elevated blood glucose levels, leads to severe disturbances in myocardial metabolism [57,58]. The administration of DOX undoubtedly further aggravates these abnormal changes in myocardial metabolism [59]. It is noteworthy that in studies focusing on diabetic vascular remodeling, Si/Cur has been shown to effectively inhibit the vicious cycle of inflammation induced by hyperglycemia by activating the Nrf2/HO-1 signaling pathway. Crucially, the Nrf2/HO-1 signaling pathway is also widely recognized as a core signaling pathway for improving myocardial metabolism and treating cardiac dysfunction [[60], [61], [62]]. Based on these studies, we made a reasonable inference: Si/Cur may also have the potential to further block DOX-mediated myocardial injury through the Nrf2/HO-1 signaling pathway. Excitingly, subsequent research confirmed this, demonstrating that Si/Cur exerts its ameliorative effects against DOX-induced myocardial toxicity through the HO-1/NRF2 pathway. This finding not only deepens our understanding of myocardial protection mechanisms but also provides new insights and potential therapeutic strategies for the prevention and treatment of cardiac toxicity associated with chemotherapeutic drugs.

Remarkably, our network pharmacological analysis in this study revealed, beyond curcumin, other polyphenolic compounds with potentially significant effects, including resveratrol, genistein, and quercetin. These compounds, exhibiting anti-inflammatory properties and myocardial tissue-protective abilities, emerge as promising contenders for the treatment of chemotherapy-induced cardiotoxicity and diabetic vascular remodeling [[63], [64], [65]]. When compared to therapies such as stem cell therapy, extracellular vesicle therapy, and growth factor therapy, these polyphenolic compounds offer inherent advantages of easy accessibility and cost-effectiveness, rendering them more suitable for promotion and application. Moving forward, we will prioritize clinical translational research on these polyphenolic compounds to better address the therapeutic requirements of patients with these complex diseases.

Although we have found that Si/Cur microspheres can effectively inhibit vascular remodeling and reduce myocardial damage by activating the Nrf2/HO-1 pathway, thereby breaking the vicious cycle of persistent cardiovascular injury, the specific biological pathways through which Si/Cur microspheres activate this pathway need further elucidation. Current research indicates that the specific structure in the Cur molecule can bind to key residues of the Keap1 protein, triggering a series of reactions that ultimately lead to the activation of Nrf2 and the transcription of antioxidant genes [37,38]. This suggests that Cur may be a direct stimulus for the Nrf2/HO-1 pathway, thereby exerting its therapeutic efficacy. It is foreseeable that when Si/Cur microspheres effectively inhibit vascular remodeling and reduce myocardial damage through the Nrf2/HO-1 pathway, they will significantly reduce the high mortality rate in these patients. Specifically, this treatment not only effectively reduces the incidence of cardiovascular events by stabilizing plaques and improving microcirculation to lower the occurrence of myocardial infarction and stroke, thereby directly reducing cardiovascular-related deaths; it also alleviates myocardial fibrosis and improves metabolism, delaying the progression of heart failure and improving patient survival rates. Additionally, the protective effect on the myocardium reduces the risk of myocarditis and cardiomyopathy induced by chemotherapeutic drugs. Improved cardiovascular function will enable patients to tolerate more intense or prolonged chemotherapy/radiation therapy, thereby enhancing cancer control rates and further reducing mortality. In summary, the Si/Cur microsphere treatment, with its dual cardiovascular protective effects, provides an effective means for treating this complex disease and brings new hope to clinical practice.

In this study, we meticulously designed two administration methods for Si/Cur: one is the directly prepared Si/Cur active solution, and the other is the Si/Cur microsphere formulation. These two administration methods each have their unique characteristics and advantages. Specifically, the Si/Cur solution can be directly administered via intravenous injection, allowing the dissolved Cur to quickly enter the bloodstream and interact more directly with the tumor-related circulatory system, thereby exerting its unique therapeutic effects [66]. This method is rapid and direct, facilitating the swift achievement of therapeutic goals. On the other hand, Si/Cur microspheres offer the significant advantage of sustained drug release [67]. Following a single administration, the microspheres can slowly release Si ions and Cur over an extended period, achieving long-lasting treatment for the disease. This approach not only reduces the inconvenience of frequent dosing but also enhances the continuity and stability of treatment. These two innovative therapeutic strategies provide new ideas and options for the chemical engineering field in heart disease treatment [68], holding promise for better treatment outcomes and quality of life for heart disease patients.

In addition to the known vascular remodeling in diabetes and chemotherapy-induced cardiotoxicity, the potential of Si/Cur microspheres in the treatment of other diseases warrants attention. Particularly, given the close correlation between the pancreas and diabetes, a metabolic disorder characterized by hyperglycemia, whose pathogenesis is closely linked to pancreatic dysfunction. Prolonged hyperglycemia may lead to inflammatory responses in the pancreas, such as autoimmune pancreatitis, further impairing pancreatic function [69,70]. Our study reveals that Si/Cur exhibits hypoglycemic effects, suggesting its potential application in diabetes treatment. Notably, previous research has shown that Si ions alone can treat chronic pancreatitis by regulating the interactions between pancreatic acinar cells, macrophages, and pancreatic stellate cells [71]. Therefore, we have reason to believe that Si/Cur microspheres may further enhance the therapeutic efficacy in pancreatic-related diseases, demonstrating potential therapeutic value especially in conditions such as pancreatic inflammation and cancer transformation.

4. Conclusion

This study integrates clinical cohort research with network pharmacology analysis, leveraging deep data mining techniques to propose innovative biomaterial design strategies for the treatment of cardiovascular diseases in the complex population of diabetic cancer patients undergoing chemotherapy. Based on this approach, we have successfully developed silicified curcumin (Si/Cur), which uses Si to create an alkaline microenvironment, facilitating efficient delivery of Cur and maximizing its biological activity. Meanwhile, the synergistic effect between Si and Cur effectively protects NRCMs from DOX-induced damage, while reducing inflammatory responses and oxidative stress behaviors triggered by high-glucose environments in endothelial cells and smooth muscle cells. This, in turn, blocks the vicious cycle of inflammation and opens up new avenues for the treatment of this complex clinical condition. This multidisciplinary treatment approach not only deeply analyzes clinical cases through big data mining but also provides innovative biomaterial design strategies for coexisting diseases that urgently require clinical attention, bringing new insights and hope to the field of disease treatment.

5. Materials and methods

5.1. Prospective cohort study

Data Source and Study Population: This study is a prospective cohort study utilizing data from the UK Biobank (UKB), which contains information on over 500,000 UK individuals and is one of the largest global repositories of human biological and health information. This study specifically focuses on patients with both diabetes and cancer who received chemotherapy (DMCA) and cancer patients without diabetes who received chemotherapy (CA), recorded in the UKB between 2006 and 2010, aiming to deeply analyze the prevalence, prognosis, and cardiovascular damage of these two types of patients.

Inclusion and Exclusion Criteria: The inclusion criteria are patients diagnosed with cancer and receiving chemotherapy between 2006 and 2010, with clear information on diabetes comorbidity. Exclusion criteria include: (1) lack of follow-up records; (2) missing key covariate information such as age, race, gender, smoking history, alcohol consumption history, BMI, education level, history of hypertension and hyperlipidemia, etc. Ethical approval for this study was provided by the UKB, and detailed ethical approval information is available in the Supplementary Methods section.

Included Variables and Endpoint Events: Patients in this study were divided into two groups: the chemotherapy cancer with diabetes group and the chemotherapy cancer without diabetes group. Covariates cover age, gender, race, smoking status, alcohol consumption status, BMI, education level, hypertension, and hyperlipidemia. Endpoint events are defined as all-cause mortality and cardiovascular disease (CVD) mortality. Among them, CVD mortality cases are defined according to the statistical classification of the International Classification of Diseases, 10th Revision (ICD-10) [72]. The follow-up period is calculated from the cancer diagnosis date between 2006 and 2010 and terminates at the date of the patient's last personal contact with the UKB or the date of all-cause mortality.

Statistical Methods: In the baseline data, categorical variables are tested using the chi-square test and expressed as frequencies and percentages; continuous variables are tested using the T-test and expressed as mean (standard deviation). This study analyzes the proportion of chemotherapy cancer patients with diabetes from 2006 to 2022 and its changes over the years. Kaplan-Meier (KM) survival curves are used to analyze the overall survival (OS) and CVD-specific survival of DMCA patients and CA patients. To further compare the OS and CVD risks between chemotherapy cancer patients with and without diabetes, this study adjusts for potential confounding factors through multivariate models. Model 1 adjusts for age, gender, and race, while Model 2 further adjusts for BMI, smoking status, alcohol consumption status, income level, education level, hypertension, and hyperlipidemia based on Model 1. To account for the potential confounding influence of the sequence of diabetes onset, we further conducted subgroup analyses, categorizing patients into cancer patients with pre-existing diabetes and cancer patients diagnosed with diabetes during follow-up. All data are analyzed statistically using R software version 4.3.3, and P < 0.05 is considered statistically significant.

5.2. Network pharmacological analysis

Screening and Identification of Cardiac Injury-Related Targets: First, the Disgenet database (https://www.disgenet.org/) was utilized to retrieve myocardial injury diseases and collect reported cardiac injury-related targets, followed by the removal of duplicate genes and false-positive genes. Given the large number of targets obtained from the initial screening, we further employed the STRING database (https://cn.string-db.org/) to construct a disease protein-protein interaction (PPI) network and set a comprehensive score threshold of >0.80 to screen out core genes as research targets.

Collection and Processing of Vascular Injury Targets: Similarly, we collected vascular injury-related targets through the Disgenet database by inputting the keyword “Vasculitis” to obtain targets related to both cardiac and vascular injuries, and then performed deduplication and removal of false-positive genes.

Drug Screening Process: The identified core targets of cardiac and vascular injuries were input into the DGIdb drug-gene interaction database (https://www.dgidb.org/) to screen for drugs with potential therapeutic effects.

Identification of Common Drugs for Cardiac and Vascular Injuries and Selection of Core Drugs: By drawing a Venn diagram, we identified drugs that target both cardiac and vascular injuries. On this basis, further screening was conducted based on scores and target numbers, and drugs with fewer than 25 targets of action were eliminated to determine the core drugs. Finally, the selected drugs and their corresponding disease targets were imported into R software, and the “drug-disease target” network diagram was drawn using the ggplot2 package to visually display the relationship between drugs and disease targets.

5.3. Preparation and characterization of Si/curcumin (Si/Cur) composite solution

The Si ion solution was prepared by calcium silicate bioceramics powder, which was soaked in various solvents, including double-distilled water, PBS, ECM basal medium, DMEM basal medium, or normal saline, at a mass to liquid volume ratio of 200 mg/mL for a duration of 24 h. During this period, the mixture was agitated at 120 rpm on a constant temperature shaker set at 37 °C to facilitate thorough interaction between the powder and the solvent. Following the soaking process, the mixture underwent centrifugation at 4000 rpm for 20 min. Afterwards, curcumin (Cur, sourced from Macklin, CAS number: 458-37-7) was mechanically blended with the Si solution. The Si/Cur mixture underwent the same extraction process as the Si solution. Following this, the blended solution was filtered through a 0.22 μm filter, yielding the Si/Cur composite solution. The solubility of Cur in this composite was determined by measuring the absorption peak intensity of the solution at 425 nm using a UV–visible spectrophotometer and comparing it to the solubility of Cur in DMSO. ABTS, H2O2 and •O2− reacted with Si/Cur and Cur and were detected by UV spectrophotometer to evaluate the free radical scavenging ability of Si/Cur. Furthermore, the free radical scavenging capacity of Si/Cur by was assessed observing its reactions with ABTS, H2O2, and •O2, which were detected using a UV spectrophotometer and compared to the reactions of Cur alone.

5.4. Design and material characterization of Si/Cur Microspheres

In this study, the sol-gel method was employed to prepare Si/Cur microspheres. The specific steps are as follows: First, 1.82 g of cetyltrimethylammonium bromide (CTAB) and 3.00 g of ammonium fluoride (NH4F) were added to a beaker containing 500 mL of deionized water and stirred at 80 °C for 1 h. Subsequently, a certain amount of calcium nitrate was dissolved in a small amount of anhydrous ethanol and treated with ultrasonication to ensure complete dissolution, followed by uniformly mixing with 9.0 mL of tetraethyl orthosilicate. Next, a 10 mL syringe was used to slowly drop the mixed sample into the above reaction solution, and the reaction was continued for 4 h. After the reaction was completed, the container was removed from the 80 °C water bath, cooled to room temperature, and allowed to stand overnight. Afterwards, the solution was centrifuged at 8000 rpm for 25 min and washed alternately with anhydrous ethanol and deionized water. Finally, the cleaned sample was freeze-dried and calcined in a muffle furnace at 600 °C for 6 h to obtain Si microspheres.

Next, 0.2 g of Si microspheres was dispersed into 100 mL of an alcoholic solution containing curcumin (Cur, 2 mg/mL). The mixed solution was stirred vigorously for 48 h to ensure that Cur was completely infiltrated into the nanoparticles. Subsequently, the mixed solution was centrifuged at 8000 rpm for 5 min, washed three times with alcohol to thoroughly remove Cur from the surface of the nanoparticles, and then freeze-dried for 24 h to finally obtain Si/Cur microspheres.

To characterize the Si/Cur microspheres, we employed a variety of advanced instruments. The morphology of the Si/Cur microspheres was observed using a scanning electron microscope (SEM, S-4800, Hitachi, Japan). Specifically, a Si/Cur microsphere concentration of 50 μg/mL was used during the SEM imaging process. The microstructure of the Si/Cur microspheres was further analyzed by a transmission electron microscope (TEM, JEM-2100F, JEOL, Japan). Simultaneously, the release concentration of SiO32− ions was determined using ICP-AES (Thermo Fisher X Series 2, USA), and the release concentration of Cur was measured using a UV–Vis spectrophotometer.

5.5. Establishment and in vivo experimentation of a mouse model of diabetes mellitus with doxorubicin-induced cardiac injury

All animal experiments in this study were approved by the Ethics Committee of the Tenth Affiliated Hospital of Southern Medical University (IACUC-AWEC-202310001). We selected db/db male diabetic mice(weighing 45 g and aged 8 weeks) with homozygous mutations in the leptin receptor gene (purchased from Changzhou Cavens Laboratory Animal Co., Ltd.) and induced doxorubicin-induced cardiac toxicity in them. According to literature reports, acute disseminated intravascular coagulation (DIC) is more common in clinical settings and predicts poor clinical outcomes [[73], [74], [75], [76]]. Therefore, we established an acute DIC model by administering a single dose of doxorubicin hydrochloride (DOX, MCE, HY-15142) (15 mg/kg, intraperitoneal injection) to the mice, while the sham group was injected with an equal volume of saline. The mice were randomly divided into groups and ensured sufficient and equal access to food and water for each group. The groups were as follows: (a) Sham group; (b) Control group (DOX); (c) Si group (DOX + Si); (d) Cur group (DOX + Cur); (e) Si/Cur group (DOX + Si/Cur).

After the doxorubicin administration for model establishment, the Si/Cur group immediately began receiving Si/Cur microsphere intervention (100 μg/mL), with each mouse receiving an intravenous injection of 8 μL/g once daily. The sham group, control group, and Si microsphere group were intravenously injected with equal volumes and concentrations of saline and Si microsphere solution, respectively, for 5 consecutive days. On the 5th day, the mice underwent echocardiography, followed by euthanasia, and samples were collected for further study, including the ratio of heart weight to tibia length, Masson's trichrome staining of heart sections, and TUNEL assay. All tissue samples underwent both tissue section staining and immunohistochemical staining, from which representative images were meticulously selected for presentation.

5.6. Echocardiography

To assess the cardiac function of mice, we employed a non-invasive transthoracic echocardiography using the Vevo 2100 system equipped with a 30-MHz transducer. The mice were first anesthetized with 5 % isoflurane. Subsequently, we obtained short-axis two-dimensional guided M-mode echocardiograms to measure the left ventricular fractional shortening (LVFS) and left ventricular ejection fraction (LVEF).

5.7. Masson's trichrome staining and TUNEL staining

Heart tissues were fixed in 4 % paraformaldehyde, embedded in paraffin, and sectioned at 5 mm. Masson's trichrome staining was used to evaluate the degree of cardiac fibrosis, and TUNEL staining was used to assess myocardial apoptosis. Image J software was utilized for data analysis of both Masson's staining and TUNEL staining.

5.8. Heart weight to tibia length ratio

Through a midline laparotomy, we performed retrograde perfusion with cold saline to clear residual blood cells in the myocardial tissue. After perfusion, the hearts of the animals were removed from the thoracic cavity, blotted dry, and promptly weighed on a scale to obtain the heart weight (HW). Simultaneously, we measured the tibia length (TL) and calculated the ratio of HW to TL.

5.9. Extraction of primary cardiomyocytes and in vitro experimentation

We obtained 1-2-day-old neonatal Sprague-Dawley (SD) rats from the Experimental Animal Center of Southern Medical University. Following a previously reported method [77], we isolated primary neonatal rat cardiomyocytes (NRCMs) from the hearts of these neonatal SD rats. For the in vitro experiment, NRCMs were randomly divided into five groups as follows: (a) Blank group; (b) Control group (DOX); (c) Si group (DOX + Si); (d) Cur group (DOX + Cur); and (e) Si/Cur group (DOX + Si/Cur). We incubated NRCMs with 0.5 μM DOX for 24 h to induce an Adriamycin (DOX) cardiomyopathy model. Among these, the Si/Cur group was treated with a Si/Cur composite solution (stock solution concentrations: Si at 120 μg/mL and Cur at 240 μg/mL) for 24 h, while the Si group and Cur group were treated with equal amounts of Si and Cur, respectively, for the same duration. Subsequently, we assayed cell viability using the CCK8 experiment and detected cardiomyocyte apoptosis using flow cytometry.

5.10. Flow Cytometric analysis of NRCMs

We used the Annexin V-FITC Apoptosis Detection Kit to analyze the apoptotic rate of cells. Under light-blocking conditions, we incubated the cells with Annexin V-FITC and PI for 10–15 min. After staining, the cells were washed twice with PBS and immediately analyzed by flow cytometry.

5.11. Evaluation of cell compatibility of Si/curcumin (Si/Cur) solution

To assess the cell compatibility of the Si/Cur composite solution, we selected human umbilical vein endothelial cells (HUVECs) at their fourth passage, which exhibited robust growth. Following digestion with 0.25 % trypsin, the cells were resuspended in culture medium, counted, and seeded into a 96-well plate at a density of 2000 cells per well with 100 μL of cell suspension per well. After a 24-h incubation, the HUVECs were cultured for an additional 48 h in endothelial cell basal medium (ECM) supplemented with the Si/Cur composite solution at various dilution gradients (stock solution concentrations: Si at 120 μg/mL and Cur at 240 μg/mL). Subsequently, cell viability was assessed using the CCK-8 assay by measuring absorbance (OD value) at 450 nm with a microplate reader.

5.12. Treatment of diabetic vascular remodeling

All animal experiments in this study were approved by the Ethics Committee of the Tenth Affiliated Hospital of Southern Medical University (IACUC-AWEC-202310001). To explore the therapeutic efficacy of the Si/Cur composite solution, we selected db/db diabetic male mice (weighing 45 g and aged 8 weeks) with homozygous mutations in the leptin receptor gene, procured from Changzhou Cavens Experimental Animal Co., Ltd. in Jiangsu Province. Normal blood glucose C57BL/6j male mice (weighing 20 g and aged 8 weeks) served as controls. The experiment comprised five groups: a normal mouse group (Sham group), a diabetic mouse group (Control group), a curcumin solution intervention group (Cur group), a Si/curcumin mixed solution intervention group (Si/Cur group), and a Si ion intervention group (Si group). Grouping was based on pre-experimental fasting blood glucose and body weight to ensure similar physiological indicators within each group. Baseline data were collected from the Sham and Control groups at the experiment's outset. Every two weeks, fasting body weight and blood glucose were measured. Upon experiment completion, mice were dissected, and samples were collected after full anesthesia [78,79]. Organs and vasculature from the aortic root to the common iliac arteries were harvested, fixed in 4 % paraformaldehyde for 24 h, or stored at −80 °C. Aortic tissues were processed for histological and immunohistochemical staining, including H&E, Masson, Victoria blue, ICAM-1, VCAM-1, and CD68 staining for further analysis. All tissue samples underwent both tissue section staining and immunohistochemical staining, from which representative images were meticulously selected for presentation.

5.13. Detection of myocardial injury markers, inflammatory factors, liver, and kidney function indicators in animal serum

Whole blood samples were allowed to clot naturally at room temperature for 40 min before being placed in a pre-cooled centrifuge set to 4 °C. The samples were then spun at 3000 rpm for 25 min. Using a pipette, the supernatant was carefully extracted, ensuring no contact with the clotted blood, and the required amount for testing was removed. The remaining serum was transferred to a fresh centrifuge tube and preserved at −80 °C to prevent repeated freezing and thawing. Subsequently, ELISA was utilized to accurately quantify the concentrations of myocardial injury markers, including Serum cardiac troponin T (cTnT), creatine kinase-MB isoenzyme (CK-MB), and lactate dehydrogenase (LDH), as well as inflammatory factors, specifically IL-1β, IL-6, and TNF-α. Additionally, key indicators of liver and kidney function, namely serum creatinine, urea nitrogen, alanine aminotransferase, and aspartate aminotransferase, were also assayed.

5.14. Detection of ICAM-1 and VCAM-1 protein expression in experimental animal vessels

After pre-cooling the homogenizer to 4 °C, an appropriate amount of lysis buffer was prepared in the ratio of RIPA: protease inhibitor: phosphatase inhibitor as 100:1:1. The animal tissues, accurately weighed on an analytical balance, were then placed in high-temperature sterilized centrifuge tubes (this step was performed on ice). The blood vessels were fully minced using scissors, and lysis buffer was added at a tissue mass to lysis buffer volume ratio of 1 mg:10 μL. Two small magnetic beads, pre-soaked in alcohol and dried, were added to each tube before homogenizing 3–5 times for 60 s each to ensure thorough tissue grinding. Post-homogenization, the samples were centrifuged at 12000 rpm for 15 min. The supernatant was meticulously collected, and a small amount was set aside for protein concentration measurement using the BCA assay. The rest of the supernatant was blended with loading buffer in a 1:4 proportion, then thermally treated at 100 °C for 10 min before being preserved at −80 °C for later use in Western blot protein electrophoresis. Western blot analysis was subsequently conducted to detect ICAM-1 and VCAM-1 proteins, with the exposed band images undergoing gray value analysis via Image J software. Lastly, comprehensive statistical analysis was undertaken to discern group disparities.

5.15. Adhesion experiment of monocyte endothelial cells (THP-1)

THP-1 cells, originating from a human monocyte line, are suspension-growing cells. These cells were cultured in a complete medium consisting of RPMI-1640, supplemented with 10 % fetal bovine serum and 1 % double antibodies. The incubation conditions were maintained at 37 °C with 5 % CO2. Healthy, proliferating THP-1 cells were selected for centrifugation and resuspension. Subsequently, 1 μL of Calcein AM was added per 1 mL of cell suspension. This dye specifically stains live cells with green fluorescence, aiding in their fluorescent labeling. The labeled cells were incubated for 30 min before being centrifuged, resuspended, and washed three times with serum-free RPMI-1640 to eliminate any residual dye that could interfere with umbilical vein endothelial cell staining. After gently washing the treated umbilical vein endothelial cells twice with PBS, RPMI-1640 complete medium-resuspended THP-1 cells were introduced and incubated for 2 h. Following incubation, the cells were observed and photographed using an inverted fluorescence microscope. Three high-power microscope fields were randomly chosen from each group for analysis, and the number of THP-1 cells in each field was tallied. The average count was used as the representative value for each group. A statistical analysis was then conducted to discern any significant differences among the groups.

5.16. Reactive oxygen species (ROS) fluorescence staining

To assess intracellular reactive oxygen species levels, we employed the fluorescent probe DCFH-DA. In our experiment, the DCFH-DA stock solution was diluted 1000-fold to a final concentration of 10 μmol/L using serum-free medium. We then added 1 mL of the diluted DCFH-DA solution to each well to cover the cells completely. After incubating the cells for 20 min in a culture incubator, we carefully washed them three times with serum-free medium to eliminate residual probes. Subsequently, the washed cells were examined and photographed under a fluorescence microscope. We randomly selected three high-power fields from each well and photographed them under consistent conditions. The fluorescence intensity of individual cells was quantified using ImagePro Plus 6 software.

5.17. Nrf2 fluorescence staining

We selected well-grown 4th-generation umbilical vein endothelial cells, digested, resuspended, and seeded them onto 24-well plates with glass slides. After allowing the cells to adhere tightly to the slides for 24 h in a culture incubator, we introduced intervention solutions, such as Si/Cur, and maintained the treatment for 48 h (replacing the solution every 24 h while avoiding PBS washes during replacement). Following the intervention, we gently removed the cell culture medium using a pipette and washed the cells twice with PBS. We then added 500 μL of 4 % paraformaldehyde to each well and fixed the cells at room temperature for 15 min. After paraformaldehyde removal and subsequent PBS washes, we diluted the Nrf2 monoclonal antibody with the primary antibody diluent at a 1:500 vol ratio. We added 200 μL of the diluted antibody to each well and incubated the cells at 4 °C for a minimum of 12 h or overnight. Subsequently, we introduced 200 μL of DAPI reagent to each well to stain the cell nuclei and incubated the cells in the dark at room temperature for 6 min. After washing with PBST, taking precautions to avoid cell detachment, we mounted the slides using an anti-fade mounting medium and observed them under a fluorescence microscope for photographic documentation.

5.18. Cell scratch assay

Once the cells (2∗106) had completely covered the bottom of the 6-well plate, we positioned a sterilized ruler on the Petri dish, ensuring it aligned perpendicularly with previously marked lines. We then made a precise scratch and replaced the original medium with a specialized intervention medium containing 1 % serum. The cells were cultured for an additional 48 h, with the intervention solution being replaced after 24 h (avoiding PBS washes during this process). Immediately after scratching, we photographed the initial state, focusing on areas of consistent width and clear visibility. The scratch area in each photograph was measured using ImageJ software to calculate the cell migration rate.

5.19. Detection of ICAM-1, VCAM-1, Nrf2, HO-1, and α-SMA protein expression

HUVECs or smooth muscle cells (SMCs) or NRCMs were carefully selected, digested, resuspended, and plated in 35 mm diameter petri dishes. After allowing the cells to adhere to the dish for 24 h, we introduced a specialized intervention medium containing 5 % fetal bovine serum and 0.2 % growth factors for a 24-h treatment. The medium was then replaced with a fresh one, and the cells were cultured for an additional 24 h and 48 h of intervention. Finally, Western blot protein electrophoresis was employed to ascertain the expression levels of ICAM-1, VCAM-1, Nrf2, HO-1, and α-SMA proteins.

5.20. Detection of inflammatory factors released by SMCs

SMCs were seeded in a 6-well plate. After being incubated for 24 h, 2.5 mL of the prepared intervention medium, enriched with 5 % fetal bovine serum, was administered to each well. Following a 24-h exposure, the cells were treated with fresh intervention medium and cultured for another 24 h, amounting to a total intervention duration of 48 h. Afterwards, we gathered the intervention fluid and culture medium from the smooth muscle cells and precisely quantified the level of the inflammatory factor IL-1β using ELISA.

5.21. Statistical analysis

Data analysis was conducted with IBM SPSS Statistic 26 software. For comparisons involving multiple groups, ANOVA analysis was used, while comparisons between two samples relied on the t-test. Statistical significance was determined at P < 0.05. All collected data were presented as mean ± standard deviation.

CRediT authorship contribution statement

Tianwang Guan: Writing – review & editing, Writing – original draft, Visualization, Methodology, Investigation, Funding acquisition, Data curation, Conceptualization. Zhenxing Lu: Writing – review & editing, Writing – original draft, Visualization, Methodology, Investigation, Formal analysis, Data curation. Rundong Tai: Writing – original draft, Visualization, Methodology, Investigation, Formal analysis. Shuai Guo: Writing – original draft, Visualization, Methodology, Investigation, Formal analysis. Zhaowenbin Zhang: Writing – review & editing, Methodology, Investigation, Formal analysis, Conceptualization. Shaohui Deng: Writing – original draft, Visualization, Investigation, Formal analysis. Jujian Ye: Writing – original draft, Visualization, Investigation, Formal analysis. Kaiyi Chi: Writing – original draft, Investigation, Data curation. Binghua Zhang: Writing – original draft, Investigation, Data curation. Huiwan Chen: Writing – original draft, Investigation, Data curation. Zhilin Deng: Writing – original draft, Investigation. Yushen Ke: Writing – original draft, Investigation. Andong Huang: Writing – original draft, Investigation. Peier Chen: Conceptualization, Formal analysis, Supervision, Writing – original draft, Writing – review & editing. Chunming Wang: Writing – review & editing, Project administration, Methodology, Conceptualization. Caiwen Ou: Writing – review & editing, Supervision, Project administration, Methodology, Funding acquisition, Conceptualization.

Data availability statement

Data will be made available on request.

Ethics approval and consent to participate

The animal study protocol was approved by the Ethics Committee of the Tenth Affiliated Hospital of Southern Medical University, protocol number IACUC-AWEC-202310001. The study adhered to the guidelines set by the committee. The cohort study utilized data from the UK Biobank Resource under Application Number 143798.

Funding sources

This work was supported by the National Natural Science Foundation of China (Grant Nos. 32371428, 82172103, 82403685 and 32301096), China Postdoctoral Science Foundation (Grant No. 2023M741567), National Key Specialist Funding Cultivation Fund (Grant No. Z202304), Guangdong Basic, Applied Basic Research Foundation (Grant Nos. 2023A1515110724, 2023B1515130005 and 2022B1515120065), Dongguan Science and Technology of Social Development Program (Grant No. G202306), and Postdoctoral Fellowship Program of CPSF (Grant No. GZC20240662).

Declaration of competing interest

Chunming Wang is an editorial board member for Bioactive Materials and was not involved in the editorial review or the decision to publish this article. All authors declare that there are no competing interests.

Acknowledgements

The cohort study was conducted using the UK Biobank resource. We thank the participants of the UK Biobank.

Footnotes

Peer review under the responsibility of KeAi Communications Co., Ltd.

Appendix A

Supplementary data to this article can be found online at https://doi.org/10.1016/j.bioactmat.2025.03.007.

Contributor Information

Zhaowenbin Zhang, Email: zhangzhaowb0058@163.com.

Peier Chen, Email: cpe0622@163.com.

Chunming Wang, Email: CMWang@umac.mo.

Caiwen Ou, Email: oucaiwen@smu.edu.cn.

Appendix A. Supplementary data

The following is the Supplementary data to this article:

Multimedia component 1
mmc1.docx (920.7KB, docx)

References

  • 1.Hossain M.J., Al‐Mamun M., Islam M.R. Diabetes mellitus, the fastest growing global public health concern: early detection should be focused. Health Sci. Rep. 2024;7(3) doi: 10.1002/hsr2.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Siegel R.L., Giaquinto A.N., Jemal A. Cancer statistics. Ca - Cancer J. Clin. 2024;74(1) doi: 10.3322/caac.21820. 2024. [DOI] [PubMed] [Google Scholar]
  • 3.Ameri P., Bertero E., Lombardi M., Porto I., Canepa M., Nohria A., Vergallo R., Lyon A.R., López-Fernández T. Ischaemic heart disease in patients with cancer. Eur. Heart J. 2024;45(14):1209–1223. doi: 10.1093/eurheartj/ehae047. [DOI] [PubMed] [Google Scholar]
  • 4.Xu Q.-Q., Han S.-J., Wei X.-H., You L.-z., Sun L.-C., Shang H.-C. Risk factors for arrhythmias occurred in cancer patients after chemotherapy: an evidence-based systematic review and meta-analysis. Heliyon. 2024;10(14) doi: 10.1016/j.heliyon.2024.e34176. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Gray S.P., Jandeleit-Dahm K. The pathobiology of diabetic vascular complications—cardiovascular and kidney disease. J. Mol. Med. 2014;92(5):441–452. doi: 10.1007/s00109-014-1146-1. [DOI] [PubMed] [Google Scholar]
  • 6.Dal Canto E., Ceriello A., Rydén L., Ferrini M., Hansen T.B., Schnell O., Standl E., Beulens J.W. Diabetes as a cardiovascular risk factor: an overview of global trends of macro and micro vascular complications. European J. Prevent. Cardiol. 2019;26(2_suppl):25–32. doi: 10.1177/2047487319878371. [DOI] [PubMed] [Google Scholar]
  • 7.Zheng P., Fan M., Liu H., Zhang Y., Dai X., Li H., Zhou X., Hu S., Yang X., Jin Y. Self-propelled and near-infrared-phototaxic photosynthetic bacteria as photothermal agents for hypoxia-targeted cancer therapy. ACS Nano. 2020;15(1):1100–1110. doi: 10.1021/acsnano.0c08068. [DOI] [PubMed] [Google Scholar]
  • 8.Tang J., Vandergriff A., Wang Z., Hensley M.T., Cores J., Allen T.A., Dinh P.-U., Zhang J., Caranasos T.G., Cheng K. A regenerative cardiac patch formed by spray painting of biomaterials onto the heart. Tissue Eng. C Methods. 2017;23(3):146–155. doi: 10.1089/ten.tec.2016.0492. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Yee K., Malliaras K., Kanazawa H., Tseliou E., Cheng K., Luthringer D.J., Ho C.-S., Takayama K., Minamino N., Dawkins J.F. Allogeneic cardiospheres delivered via percutaneous transendocardial injection increase viable myocardium, decrease scar size, and attenuate cardiac dilatation in porcine ischemic cardiomyopathy. PLoS One. 2014;9(12) doi: 10.1371/journal.pone.0113805. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Cheng K., Kisaalita W.S. Exploring cellular adhesion and differentiation in a micro‐/nano‐hybrid polymer scaffold. Biotechnol. Prog. 2010;26(3):838–846. doi: 10.1002/btpr.391. [DOI] [PubMed] [Google Scholar]
  • 11.Cheng K., Malliaras K., Shen D., Tseliou E., Ionta V., Smith J., Galang G., Sun B., Houde C., Marbán E. Intramyocardial injection of platelet gel promotes endogenous repair and augments cardiac function in rats with myocardial infarction. J. Am. Coll. Cardiol. 2012;59(3):256–264. doi: 10.1016/j.jacc.2011.10.858. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Xie Y., Ibrahim A., Cheng K., Wu Z., Liang W., Malliaras K., Sun B., Liu W., Shen D., Cheol Cho H. Importance of cell-cell contact in the therapeutic benefits of cardiosphere-derived cells. Stem Cell. 2014;32(9):2397–2406. doi: 10.1002/stem.1736. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Rahman S., Rahman T., Ismail A.A.S., Rashid A.R.A. Diabetes‐associated macrovasculopathy: pathophysiology and pathogenesis, Diabetes. Obes. Metabol. 2007;9(6):767–780. doi: 10.1111/j.1463-1326.2006.00655.x. [DOI] [PubMed] [Google Scholar]
  • 14.Toma L., Stancu C.S., Sima A.V. Endothelial dysfunction in diabetes is aggravated by glycated lipoproteins; novel molecular therapies. Biomedicines. 2020;9(1):18. doi: 10.3390/biomedicines9010018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Méndez-Barbero N., Gutiérrez-Muñoz C., Blanco-Colio L.M. Cellular crosstalk between endothelial and smooth muscle cells in vascular wall remodeling. Int. J. Mol. Sci. 2021;22(14):7284. doi: 10.3390/ijms22147284. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Zhang X., Hu C., Kong C.-Y., Song P., Wu H.-M., Xu S.-C., Yuan Y.-P., Deng W., Ma Z.-G., Tang Q.-Z. FNDC5 alleviates oxidative stress and cardiomyocyte apoptosis in doxorubicin-induced cardiotoxicity via activating AKT. Cell Death Differ. 2020;27(2):540–555. doi: 10.1038/s41418-019-0372-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Arola O.J., Saraste A., Pulkki K., Kallajoki M., Parvinen M., Voipio-Pulkki L.-M. Acute doxorubicin cardiotoxicity involves cardiomyocyte apoptosis. Cancer Res. 2000;60(7):1789–1792. [PubMed] [Google Scholar]
  • 18.Hosseini M.S., Bejnordi B.E., Trinh V.Q.-H., Chan L., Hasan D., Li X., Yang S., Kim T., Zhang H., Wu T. Computational pathology: a survey review and the way forward. J. Pathol. Inf. 2024;15 doi: 10.1016/j.jpi.2023.100357. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Guo X., Luo W., Wu L., Zhang L., Chen Y., Li T., Li H., Zhang W., Liu Y., Zheng J. Natural products from herbal medicine self‐assemble into advanced bioactive materials. Adv. Sci. 2024;11(35) doi: 10.1002/advs.202403388. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Henrotin Y., Priem F., Mobasheri A. Curcumin: a new paradigm and therapeutic opportunity for the treatment of osteoarthritis: curcumin for osteoarthritis management. SpringerPlus. 2013;2:1–9. doi: 10.1186/2193-1801-2-56. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Li Y., Jiao J., Qi Y., Yu W., Yang S., Zhang J., Zhao J. Curcumin: a review of experimental studies and mechanisms related to periodontitis treatment. J. Periodontal. Res. 2021;56(5):837–847. doi: 10.1111/jre.12914. [DOI] [PubMed] [Google Scholar]
  • 22.Mito S., Watanabe K., Harima M., Thandavarayan R.A., Veeraveedu P.T., Sukumaran V., Suzuki K., Kodama M., Aizawa Y. Curcumin ameliorates cardiac inflammation in rats with autoimmune myocarditis. Biol. Pharm. Bull. 2011;34(7):974–979. doi: 10.1248/bpb.34.974. [DOI] [PubMed] [Google Scholar]
  • 23.Schiborr C., Kocher A., Behnam D., Jandasek J., Toelstede S., Frank J. The oral bioavailability of curcumin from micronized powder and liquid micelles is significantly increased in healthy humans and differs between sexes. Mol. Nutr. Food Res. 2014;58(3):516–527. doi: 10.1002/mnfr.201300724. [DOI] [PubMed] [Google Scholar]
  • 24.Priyadarsini K.I. The chemistry of curcumin: from extraction to therapeutic agent. Molecules. 2014;19(12):20091–20112. doi: 10.3390/molecules191220091. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Gao Z., Yang X., Huang K., Xu H. Free-radical scavenging and mechanism study of flavonoids extracted from the radix of Scutellaria baicalensis Georgi. Appl. Magn. Reson. 2000;19(1):35–44. doi: 10.1016/s0304-4165(99)00152-x. [DOI] [PubMed] [Google Scholar]
  • 26.Jurasekova Z., Domingo C., Garcia-Ramos J.V., Sanchez-Cortes S. Effect of pH on the chemical modification of quercetin and structurally related flavonoids characterized by optical (UV-visible and Raman) spectroscopy. Phys. Chem. Chem. Phys. 2014;16(25):12802–12811. doi: 10.1039/c4cp00864b. [DOI] [PubMed] [Google Scholar]
  • 27.Yuan Y., Zhang Z., Mo F., Yang C., Jiao Y., Wang E., Zhang Y., Lin P., Hu C., Fu W., Chang J., Wang L. A biomaterial-based therapy for lower limb ischemia using Sr/Si bioactive hydrogel that inhibits skeletal muscle necrosis and enhances angiogenesis. Bioact. Mater. 2023;26:264–278. doi: 10.1016/j.bioactmat.2023.02.027. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Yu Q., Han Y., Wang X., Qin C., Zhai D., Yi Z., Chang J., Xiao Y., Wu C. Copper silicate hollow microspheres-incorporated scaffolds for chemo-photothermal therapy of melanoma and tissue healing. ACS Nano. 2018;12(3):2695–2707. doi: 10.1021/acsnano.7b08928. [DOI] [PubMed] [Google Scholar]
  • 29.Wang X.T., Wang L.Y., Wu Q., Bao F., Yang H.T., Qiu X.Z., Chang J. Chitosan/calcium silicate cardiac patch stimulates cardiomyocyte activity and myocardial performance after infarction by synergistic effect of bioactive ions and aligned nanostructure. ACS Appl. Mater. Interfaces. 2019;11(1):1449–1468. doi: 10.1021/acsami.8b17754. [DOI] [PubMed] [Google Scholar]
  • 30.Srinath P., Azeem P.A., Reddy K.V. Review on calcium silicate-based bioceramics in bone tissue engineering. Int. J. Appl. Ceram. Technol. 2020;17(5):2450–2464. [Google Scholar]
  • 31.Wang B., Hu S., Teng Y., Chen J., Wang H., Xu Y., Wang K., Xu J., Cheng Y., Gao X. Current advance of nanotechnology in diagnosis and treatment for malignant tumors. Signal Transduct. Targeted Ther. 2024;9(1):200. doi: 10.1038/s41392-024-01889-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Liu X., Chen B., Chen J., Wang X., Dai X., Li Y., Zhou H., Wu L.M., Liu Z., Yang Y. A cardiac‐targeted nanozyme interrupts the inflammation‐free radical cycle in myocardial infarction. Adv. Mater. 2024;36(2) doi: 10.1002/adma.202308477. [DOI] [PubMed] [Google Scholar]
  • 33.Guan T., Jiang Y., Luo Z., Liang Y., Feng M., Lu Z., Yi M., Teng Y., Zhou R., Zeng L., Chi K., Ou C., Chen M. Long-term risks of cardiovascular death in a population-based cohort of 1,141,675 older patients with cancer. Age Ageing. 2023;52(5) doi: 10.1093/ageing/afad068. [DOI] [PubMed] [Google Scholar]
  • 34.Guan T., Zhang H., Lin W., Qiu Z., Yang J., Su M., Lai Y., Li Y., Liu C. Prognostic impacts of tumour size on patients with primary cardiac sarcoma. Can. J. Cardiol. 2020;36(8):1228–1235. doi: 10.1016/j.cjca.2019.11.033. [DOI] [PubMed] [Google Scholar]
  • 35.Guo L., Zhen Q., Zhen X., Cui Z., Jiang C., Zhang Q., Gao K., Luan D., Zhou X. A network pharmacology approach to explore and validate the potential targets of ginsenoside on osteoporosis. Int. J. Immunopathol. Pharmacol. 2022;36 doi: 10.1177/03946320221107239. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Zhang Z., Chang D., Zeng Z., Xu Y., Yu J., Fan C., Yang C., Chang J. CuCS/Cur composite wound dressings promote neuralized skin regeneration by rebuilding the nerve cell" factory" in deep skin burns. Mater. Today Bio. 2024;26 doi: 10.1016/j.mtbio.2024.101075. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Shahcheraghi S.H., Salemi F., Peirovi N., Ayatollahi J., Alam W., Khan H., Saso L. Nrf2 regulation by curcumin: molecular aspects for therapeutic prospects. Molecules. 2021;27(1):167. doi: 10.3390/molecules27010167. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Rahban M., Habibi-Rezaei M., Mazaheri M., Saso L., Moosavi-Movahedi A.A. Anti-viral potential and modulation of Nrf2 by curcumin: pharmacological implications. Antioxidants. 2020;9(12):1228. doi: 10.3390/antiox9121228. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Chou W.y.S., Hamel L.M., Thai C.L., Debono D., Chapman R.A., Albrecht T.L., Penner L.A., Eggly S. Discussing prognosis and treatment goals with patients with advanced cancer: a qualitative analysis of oncologists' language. Health Expect. 2017;20(5):1073–1080. doi: 10.1111/hex.12549. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Gramling R., Fiscella K., Xing G., Hoerger M., Duberstein P., Plumb S., Mohile S., Fenton J.J., Tancredi D.J., Kravitz R.L. Determinants of patient-oncologist prognostic discordance in advanced cancer. JAMA Oncol. 2016;2(11):1421–1426. doi: 10.1001/jamaoncol.2016.1861. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Rodbard H.W., Jellinger P.S., Davidson J.A., Einhorn D., Garber A.J., Grunberger G., Handelsman Y., Horton E.S., Lebovitz H., Levy P. Statement by an American Association of Clinical Endocrinologists/American College of Endocrinology consensus panel on type 2 diabetes mellitus: an algorithm for glycemic control. Endocr. Pract. 2009;15(6):540–559. doi: 10.4158/EP.15.6.540. [DOI] [PubMed] [Google Scholar]
  • 42.Bao Z., Li L., Geng Y., Yan J., Dai Z., Shao C., Sun Z., Jing L., Pang Q., Zhang L. Advanced glycation end products induce vascular smooth muscle cell-derived foam cell formation and transdifferentiate to a macrophage-like state. Mediat. Inflamm. 2020;2020 doi: 10.1155/2020/6850187. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Allahverdian S., Chehroudi A.C., McManus B.M., Abraham T., Francis G.A. Contribution of intimal smooth muscle cells to cholesterol accumulation and macrophage-like cells in human atherosclerosis. Circulation. 2014;129(15):1551–1559. doi: 10.1161/CIRCULATIONAHA.113.005015. [DOI] [PubMed] [Google Scholar]
  • 44.Burger F., Baptista D., Roth A., da Silva R.F., Montecucco F., Mach F., Brandt K.J., Miteva K. NLRP3 inflammasome activation controls vascular smooth muscle cells phenotypic switch in atherosclerosis. Int. J. Mol. Sci. 2021;23(1):340. doi: 10.3390/ijms23010340. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Vendrov A.E., Sumida A., Canugovi C., Lozhkin A., Hayami T., Madamanchi N.R., Runge M.S. NOXA1-dependent NADPH oxidase regulates redox signaling and phenotype of vascular smooth muscle cell during atherogenesis. Redox Biol. 2019;21 doi: 10.1016/j.redox.2018.11.021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Piconi L., Quagliaro L., Da Ros R., Assaloni R., Giugliano D., Esposito K., Szabo C., Ceriello A. Intermittent high glucose enhances ICAM‐1, VCAM‐1, E‐selectin and interleukin‐6 expression in human umbilical endothelial cells in culture: the role of poly (ADP‐ribose) polymerase. J. Thromb. Haemostasis. 2004;2(8):1453–1459. doi: 10.1111/j.1538-7836.2004.00835.x. [DOI] [PubMed] [Google Scholar]
  • 47.Cook-Mills J.M., Marchese M.E., Abdala-Valencia H. Vascular cell adhesion molecule-1 expression and signaling during disease: regulation by reactive oxygen species and antioxidants. Antioxidants Redox Signal. 2011;15(6):1607–1638. doi: 10.1089/ars.2010.3522. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Yuan T., Yang T., Chen H., Fu D., Hu Y., Wang J., Yuan Q., Yu H., Xu W., Xie X. New insights into oxidative stress and inflammation during diabetes mellitus-accelerated atherosclerosis. Redox Biol. 2019;20:247–260. doi: 10.1016/j.redox.2018.09.025. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Zhu L.-H., Wang L., Wang D., Jiang H., Tang Q.-Z., Yan L., Bian Z.-Y., Wang X.-A., Li H. Puerarin attenuates high-glucose-and diabetes-induced vascular smooth muscle cell proliferation by blocking PKCβ2/Rac1-dependent signaling. Free Radic. Biol. Med. 2010;48(4):471–482. doi: 10.1016/j.freeradbiomed.2009.10.040. [DOI] [PubMed] [Google Scholar]
  • 50.Zhang X., Liu L., Chen C., Chi Y.-L., Yang X.-Q., Xu Y., Li X.-T., Guo S.-L., Xiong S.-H., Shen M.-R. Interferon regulatory factor-1 together with reactive oxygen species promotes the acceleration of cell cycle progression by up-regulating the cyclin E and CDK2 genes during high glucose-induced proliferation of vascular smooth muscle cells. Cardiovasc. Diabetol. 2013;12:1–13. doi: 10.1186/1475-2840-12-147. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Xi G., Shen X., Wai C., White M.F., Clemmons D.R. Hyperglycemia induces vascular smooth muscle cell dedifferentiation by suppressing insulin receptor substrate-1–mediated p53/KLF4 complex stabilization. J. Biol. Chem. 2019;294(7):2407–2421. doi: 10.1074/jbc.RA118.005398. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Basatemur G.L., Jørgensen H.F., Clarke M.C., Bennett M.R., Mallat Z. Vascular smooth muscle cells in atherosclerosis. Nat. Rev. Cardiol. 2019;16(12):727–744. doi: 10.1038/s41569-019-0227-9. [DOI] [PubMed] [Google Scholar]
  • 53.Casella S., Bielli A., Mauriello A., Orlandi A. Molecular pathways regulating macrovascular pathology and vascular smooth muscle cells phenotype in type 2 diabetes. Int. J. Mol. Sci. 2015;16(10):24353–24368. doi: 10.3390/ijms161024353. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Shi L., Ji Y., Jiang X., Zhou L., Xu Y., Li Y., Jiang W., Meng P., Liu X. Liraglutide attenuates high glucose-induced abnormal cell migration, proliferation, and apoptosis of vascular smooth muscle cells by activating the GLP-1 receptor, and inhibiting ERK1/2 and PI3K/Akt signaling pathways. Cardiovasc. Diabetol. 2015;14:1–13. doi: 10.1186/s12933-015-0177-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Swamy A.V., Gulliaya S., Thippeswamy A., Koti B.C., Manjula D.V. Cardioprotective effect of curcumin against doxorubicin-induced myocardial toxicity in albino rats. Indian J. Pharmacol. 2012;44(1):73–77. doi: 10.4103/0253-7613.91871. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Licata S., Saponiero A., Mordente A., Minotti G. Doxorubicin metabolism and toxicity in human myocardium: role of cytoplasmic deglycosidation and carbonyl reduction. Chem. Res. Toxicol. 2000;13(5):414–420. doi: 10.1021/tx000013q. [DOI] [PubMed] [Google Scholar]
  • 57.Yuan S., Gill D., Giovannucci E.L., Larsson S.C. Obesity, type 2 diabetes, lifestyle factors, and risk of gallstone disease: a Mendelian randomization investigation. Clin. Gastroenterol. Hepatol. 2022;20(3):e529–e537. doi: 10.1016/j.cgh.2020.12.034. [DOI] [PubMed] [Google Scholar]
  • 58.Abel E.D. Myocardial insulin resistance and cardiac complications of diabetes. Curr. Drug Targets - Immune, Endocr. Metab. Disord. 2005;5(2):219–226. doi: 10.2174/1568008054064869. [DOI] [PubMed] [Google Scholar]
  • 59.Wu R., Wang H.-L., Yu H.-L., Cui X.-H., Xu M.-T., Xu X., Gao J.-P. Doxorubicin toxicity changes myocardial energy metabolism in rats. Chem. Biol. Interact. 2016;244:149–158. doi: 10.1016/j.cbi.2015.12.010. [DOI] [PubMed] [Google Scholar]
  • 60.Jin X., Xu Z., Cao J., Yan R., Xu R., Ran R., Ma Y., Cai W., Fan R., Zhang Y. HO-1/EBP interaction alleviates cholesterol-induced hypoxia through the activation of the AKT and Nrf2/mTOR pathways and inhibition of carbohydrate metabolism in cardiomyocytes. Int. J. Mol. Med. 2017;39(6):1409–1420. doi: 10.3892/ijmm.2017.2979. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Raish M., Ahmad A., Jardan Y.A.B., Shahid M., Alkharfy K.M., Ahad A., Ansari M.A., Abdelrahman I.A., Al-Jenoobi F.I. Sinapic acid ameliorates cardiac dysfunction and cardiomyopathy by modulating NF-κB and Nrf2/HO-1 signaling pathways in streptozocin induced diabetic rats. Biomed. Pharmacother. 2022;145 doi: 10.1016/j.biopha.2021.112412. [DOI] [PubMed] [Google Scholar]
  • 62.Hu T., Wei G., Xi M., Yan J., Wu X., Wang Y., Zhu Y., Wang C., Wen A. Synergistic cardioprotective effects of Danshensu and hydroxysafflor yellow A against myocardial ischemia-reperfusion injury are mediated through the Akt/Nrf2/HO-1 pathway Corrigendum in/10.3892/ijmm. 2021.5045. Int. J. Mol. Med. 2016;38(1):83–94. doi: 10.3892/ijmm.2016.2584. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Zhang X., Li Z., Yang P., Duan G., Liu X., Gu Z., Li Y. Polyphenol scaffolds in tissue engineering. Mater. Horiz. 2021;8(1):145–167. doi: 10.1039/d0mh01317j. [DOI] [PubMed] [Google Scholar]
  • 64.Caro C., Pourmadadi M., Eshaghi M.M., Rahmani E., Shojaei S., Paiva-Santos A.C., Rahdar A., Behzadmehr R., García-Martín M.L., Díez-Pascual A.M. Nanomaterials loaded with Quercetin as an advanced tool for cancer treatment. J. Drug Deliv. Sci. Technol. 2022;78 [Google Scholar]
  • 65.Li Z., Chen Z., Chen H., Chen K., Tao W., Ouyang X.-k., Mei L., Zeng X. Polyphenol-based hydrogels: pyramid evolution from crosslinked structures to biomedical applications and the reverse design. Bioact. Mater. 2022;17:49–70. doi: 10.1016/j.bioactmat.2022.01.038. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Allen T.A., Asad D., Amu E., Hensley M.T., Cores J., Vandergriff A., Tang J., Dinh P.-U., Shen D., Qiao L. Circulating tumor cells exit circulation while maintaining multicellularity, augmenting metastatic potential. J. Cell Sci. 2019;132(17):jcs231563. doi: 10.1242/jcs.231563. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Cheng K., Shen D., Smith J., Galang G., Sun B., Zhang J., Marbán E. Transplantation of platelet gel spiked with cardiosphere-derived cells boosts structural and functional benefits relative to gel transplantation alone in rats with myocardial infarction. Biomaterials. 2012;33(10):2872–2879. doi: 10.1016/j.biomaterials.2011.12.040. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Li Z., Hu S., Cheng K. Chemical engineering of cell therapy for heart diseases. Accounts Chem. Res. 2019;52(6):1687–1696. doi: 10.1021/acs.accounts.9b00137. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Hart P.A., Bradley D., Conwell D.L., Dungan K., Krishna S.G., Wyne K., Bellin M.D., Yadav D., Andersen D.K., Serrano J. Diabetes following acute pancreatitis. Lancet Gastroenterol.; Hepatol. 2021;6(8):668–675. doi: 10.1016/S2468-1253(21)00019-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Davison L. Diabetes mellitus and pancreatitis–cause or effect? J. Small Anim. Pract. 2015;56(1):50–59. doi: 10.1111/jsap.12295. [DOI] [PubMed] [Google Scholar]
  • 71.Yin H., Zhang Z., Zhang D., Peng L., Xia C., Yang X., Wang X., Li Z., Chang J., Huang H. A new method for treating chronic pancreatitis and preventing fibrosis using bioactive calcium silicate ion solution. J. Mater. Chem. B. 2023;11(38):9163–9178. doi: 10.1039/d3tb01287e. [DOI] [PubMed] [Google Scholar]
  • 72.Winters-Miner L.A., Bolding P.S., Hilbe J.M., Goldstein M., Hill T., Nisbet R., Walton N., Miner G.D. In: Practical Predictive Analytics and Decisioning Systems for Medicine. Winters-Miner L.A., Bolding P.S., Hilbe J.M., Goldstein M., Hill T., Nisbet R., Walton N., Miner G.D., editors. 2015. Chapter 8 - icd-10; pp. 106–115. [Google Scholar]
  • 73.Chen D., Yu W., Zhong C., Hong Q., Huang G., Que D., Wang Y., Yang Y., Rui B., Zhuang Z., Liang M., Ye Z., Yan X., Lv J., Zhang R., Yan J., Yang P. Elabela ameliorates doxorubicin-induced cardiotoxicity by promoting autophagic flux through TFEB pathway. Pharmacol. Res. 2022;178 doi: 10.1016/j.phrs.2022.106186. [DOI] [PubMed] [Google Scholar]
  • 74.Cardinale D., Sandri M.T., Martinoni A., Tricca A., Civelli M., Lamantia G., Cinieri S., Martinelli G., Cipolla C.M., Fiorentini C. Left ventricular dysfunction predicted by early troponin I release after high-dose chemotherapy. J. Am. Coll. Cardiol. 2000;36(2):517–522. doi: 10.1016/s0735-1097(00)00748-8. [DOI] [PubMed] [Google Scholar]
  • 75.Shen D., Cheng K., Marbán E. Dose‐dependent functional benefit of human cardiosphere transplantation in mice with acute myocardial infarction. J. Cell Mol. Med. 2012;16(9):2112–2116. doi: 10.1111/j.1582-4934.2011.01512.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Liu M., Hu S., Yan N., Popowski K.D., Cheng K. Inhalable extracellular vesicle delivery of IL-12 mRNA to treat lung cancer and promote systemic immunity. Nat. Nanotechnol. 2024;19(4):565–575. doi: 10.1038/s41565-023-01580-3. [DOI] [PubMed] [Google Scholar]
  • 77.Yi M., Li H., Wong X., Yan J., Gao L., He Y., Zhong X., Cai Y., Feng W., Wen Z., Wu C., Ou C., Chang J., Chen M. Ion therapy: a novel strategy for acute myocardial infarction. Adv. Sci. 2019;6(1) doi: 10.1002/advs.201801260. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Allen T.A., Gracieux D., Talib M., Tokarz D.A., Hensley M.T., Cores J., Vandergriff A., Tang J., de Andrade J.B., Dinh P.-U. Angiopellosis as an alternative mechanism of cell extravasation. Stem Cell. 2017;35(1):170–180. doi: 10.1002/stem.2451. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Li Y., Liu Y., Liu S., Gao M., Wang W., Chen K., Huang L., Liu Y. Diabetic vascular diseases: molecular mechanisms and therapeutic strategies. Signal Transduct. Targeted Ther. 2023;8(1):152. doi: 10.1038/s41392-023-01400-z. [DOI] [PMC free article] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

Multimedia component 1
mmc1.docx (920.7KB, docx)

Data Availability Statement

Data will be made available on request.


Articles from Bioactive Materials are provided here courtesy of KeAi Publishing

RESOURCES