Abstract
Vascular injury is a common complication of type 2 diabetes mellitus (T2DM). Platelet endothelial cell adhesion molecule-1 (PECAM-1) is a vascular regulator. This study is to explore the possible pathological mechanism of PECAM-1 in vascular injury in T2DM. Plasma PECAM-1 was detected using ELISA in plasma samples of T2DMs and normal subjects. NetworkAnalyst was used to analyze the PECAM-1 transcript genes. PECAM-1 transcriptional gene variation in T2DM was analyzed from GSE26168 data from the GEO database. STRING line network database was used to obtain the proteins related to PECAM-1, and the ClusterProfiler package in R language was applied to perform PPI, GO and KEGG enrichment analysis. PECAM-1targeted drugs prediction was performed by Drugbank. Compared with 66 healthy controls, the plasma PECAM-1 levels in 66 patients with T2DM were significantly decreased (pā<ā0.001). Moreover, multivariate regression analysis indicated that PECAM-1 was an independent risk factor for vascular injury in T2DM patients. GSE26168 data of T2DM blood mRNA showed that the levels of the PECAM-1 gene transcription factors CREB3, GATAD1 and TEAD3 were significantly reduced, while CUX1 and RELA were significantly increased in T2DM patients. Functional enrichment analysis of PPI, GO and KEGG suggested that PECAM-1 was involved in regulation of vascular stability, endothelial function, and angiogenesis. DrugBank search revealed that fostamatinib is a targeted drug closely matching the PECAM-1 molecule. In patients with T2DM, the decrease in PECAM-1 is an independent risk factor for vascular injury. Abnormalities in PECAM-1 transcriptional factors are likely associated with the reduction in plasma PECAM-1 levels, which may be involved in the mechanism of vascular injury in T2DM. Fostamatinib may be a candidate drug for vascular injury in T2DMs.
Similar content being viewed by others
Introduction
Diabetes has become a global epidemic disorder and the number of people with diabetes has been steadily increasing. Of these, type 2 diabetes mellitus (T2DM) accounts for the majority of diabetes cases. In 1985, only about 30Ā million people worldwide suffered from diabetes, however, by 2015, the number of the patients had increased to 392Ā million. The International Diabetes Federation estimates that approximately 463Ā million adults (ages 20ā79) worldwide have diabetes in 2019. Thatās about 6% of the worldās population. Chronic complications of diabetes are mainly related to lesions caused by microvascular injury, such as retinopathy, renal disease, cardiovascular disease and nerve damage (including peripheral nerve and autonomic neuropathy). Retinopathy affects approximately one-third of diabetic patients and is an important cause of visual impairment and blindness. Approximately 25% of T2DM patients suffer from diabetic nephropathy and end-stage renal disease. Nearly 50% of people with diabetes have diabetic neuropathy. The prevalence of erectile dysfunction in diabetic men ranges from 35 to 90%. Diabetic foot complications, including foot ulcers and amputations, are all the problems caused by microvascular injury1,2,3,4.
Vascular injury in diabetes is largely due to a combination of metabolic abnormalities, chronic hyperglycemia (high blood sugar levels), and related disturbances of various physiological pathways. The pathogenesis of diabetic vascular injury involves multiple mechanisms including: endothelial dysfunction, oxidative stress, inflammation, advanced glycation end products (AGE), increased platelet activation, dyslipidemia, atherosclerotic plaque formation, microvascular complications, impaired angiogenesis and wound healing. All of these mechanisms can cause damage to tiny blood vessels, leading to a series of vascular injury diseases5. Diabetic retinal, renal and neurological complications are usually late manifestations of microvascular injury that begins years earlier and is often influenced by other cardiometabolic disease factors (e.g. hypertension, obesity, dyslipidemia). Microvascular dysfunction in heart, muscle, and brain precedes anatomical microvascular disease. Early on, diabetes and/or cardiometabolic disease can cause reversible microvascular injury with dysfunction, which may or may not become irreversible and anatomically visible over time (e.g., Thickening of vascular basement membrane, sparse capillaries, loss of pericytes, etc.). Consequences may include common vision loss, renal insufficiency, and neuropathy, as well as escalating heart failure, sarcopenia, cognitive impairment, and metabolic dysfunction5,6. Platelet endothelial cell adhesion molecule-1 (PECAM-1), also known as CD31, is an important component of endothelial cell junctions and plays a key role in mediating cell-cell interactions, especially between endothelial cells and platelets effect. It is expressed on the surface of platelets, leukocytes and endothelial cells. PECAM-1 is involved in a variety of physiological and pathological processes, including leukocyte migration, platelet aggregation, inflammation and angiogenesis. PECAM-1 has been shown to inhibit endothelial cell proliferation, migration and tube formation, processes that are important in angiogenesis. It plays a role in regulating angiogenesis (the formation of new blood vessels) and endothelial cell function, helping maintain endothelial barrier integrity. PECAM-1 has a protective role in certain pathological conditions such as inflammation and atherosclerosis by maintaining endothelial barrier function and limiting vascular leakage7,8,9.
Based on the above background data, we hypothesize that the abnormal expression of PECAM-1 in T2DM patients may be one of the potential pathological mechanisms causing T2DM vascular injury. To this end, we first collected the baseline information and plasma samples from T2DM patients and healthy controls, detected the plasma PECAM-1 levels. The results of the multivariate logistic regression model revealed that PECAM-1 was an independent risk factor for vascular injury in T2DM patients. Further, bioinformatics analysis suggested that the alteration of PECAM-1 transcription factors, CREB3, TEAD3, GATAD1, RELA, and CUX1, may be the mechanism of T2DM plasma PECAM-1 reduction. Finally, network pharmacological analysis indicated that fostamatinib is a drug with high affinity for PECAM-1. This discovery provides novel insights into the pathological mechanism of T2DM and also identifies a new drug target for the treatment of T2DM.
Materials and methods
T2DM patients and health controls
Plasma samples were collected from 66 T2DM patients who were admitted to the Endocrinology Department of Putian University Affiliated Hospital from February 2021 to January 2023. Plasma samples were also obtained from 66 healthy adults who participated in outpatient adult health exams. Briefly, in the morning, a 5 ml forearm blood was drawn into a tube with sodium citrate anticoagulant from both the T2DM patients and the healthy controls. The tubes were then spun at 3000 x g for 20 min. The yellow supernatant plasma was collected and stored in a -80 °C freezer until it was used for ELISA assay. This study was reviewed and approved by the Ethics Committee of Affiliated Hospital of Putian University (approval number: 2022043). All patients and healthy controls participating in the survey agreed and signed informed consent forms, agreeing to provide plasma samples. This study is carried out in full accordance with the Declaration of Helsinki.
T2DM diagnostic criteria and exclusion criteria
In this study, the diagnostic criteria for T2DM were based on the definition provided by the World Health Organization (WHO)10. Patients who met the following criteria were diagnosed with T2DM and included in the study:
-
1.
Patients with symptoms of diabetes, such as polydipsia, polyphagia, polyuria, and unexplained weight loss caused by hyperglycemia, with a random blood glucose level of ā„ā11.1 mmol/L (200Ā mg/dL).
-
2.
Or, patients with symptoms of diabetes, as mentioned above, along with a fasting blood glucose level of ā„ā7 mmol/L (126Ā mg/dL).
-
3.
Or, patients with symptoms of diabetes, as mentioned above, along with a blood sugar level of ā„ā11.1 mmol/L (200Ā mg/dL) two hours after a 75Ā g glucose tolerance test.
-
4.
Or, patients with symptoms of diabetes, as mentioned above, along with a glycosylated hemoglobin (HbA1c) level of ā„ā6.5 DCCT%.
Patients without typical symptoms of diabetes were required to undergo rechecking and confirmation on another day.
Diagnostic criteria for vascular injury
-
1.
Detection via vascular ultrasound reveals an increase in the intima-media thickness (IMT) of blood vessels (carotid artery IMTāā„ā1.0Ā mm or femoral artery IMTāā„ā1.2Ā mm) or the presence of atherosclerotic plaques.
-
2.
Or, there is a decrease in the blood flow velocity in the main renal artery and its branches, along with a reduction in the peak systolic velocity (PSV), accompanied by a urinary microalbumin-to-creatinine ratio (UACR)āā„ā30Ā mg/g or a serum creatinine level elevated beyond the normal reference range11,12.
The exclusion criteria for this study were as follows: (1) Pregnancy-related diabetes and other specific types of diabetes; (2) Secondary diabetes; (3) Acute complications of diabetes observed during hospitalization; (4) Severe complications, including concurrent tumors, infections, and multiple organ failure; (5) Missing data; (6) Presence of mental illnesses such as depression, mania, and anxiety; (7) Patients who did not sign the informed consent form.
ELISA assay
The level of plasma PECAM-1 was analyzed using a Human CD31 ELISA Kit (Cat.No.: JN23015) from Jining Biotechnology (Shanghai, China), by following the manufacturerās instructions. For each sample, 20 µL of plasma was diluted to 10, 40, or 160 times, respectively, and underwent the steps of the manufactureās instruction to detect PECAM-1. The optical density (OD) value of each well at 450Ā nm was then examined by a Multiscan Spectrum (Thermo Fisher Scientific, Waltham, MA, USA). Based on a standard PECAM-1 curve, the concentration of PECAM-1 was calculated from the OD value. Each sample was repeat for three times.
Transcription factor analysis by networkanalyst
Using NetworkAnalyst (https://www.networkanalyst.ca/NetworkAnalyst/) analysis was applied to find out the PECAM-1 gene transcription factors. Based on the ENCODE database source, peak intensity signalā<ā500 and predicted regulatory potential scoreā<ā1 were used as screening coefficients to identify transcription factors related to PECAM-1 regulation.
Analysis of deferential expressed genes in T2DM blood mRNA from GEO database
Based on GEO (https://www.ncbi.nlm.nih.gov/geo/) database, a blood mRNA data of T2DM (GSE26168) was obtained, which comes from gene chip data from the Illumina HumanRef-8 v3.0 expression platform. A total of 8 control and 9 T2DM blood samples are included. The expression level of the PECAM-1 transcription factors found in NetworkAnalyst analysis were then obtained from the GSE26168 dataset (Fig.Ā 2). All significantly up-regulated and down-regulated genes were represented by a volcanic map (Fig.Ā 3A). Top 20 up-regulated and down-regulated proteins in the T2DMs were listed in a heat map.
The PECAM-1 interaction networks and function enrichment analysis
The proteins related to PECAM-1 function were analyzed using an online network database STRING (https://string-db.org/). The PECAM-1 function and interacting proteins in the human body was analyzed using nodes in the protein protein interaction (PPI) network (Fig.Ā 4A). Using the ClusterProfiler package in R language, the PECAM-1 functional related genes were divided into three parts through GO (Gene Ontology): cellular component (CC), molecular function (MF), and biological process (BP) (Fig.Ā 4B). KEGG (Kyoto Encyclopedia of Genes and Genomes) was employed to annotate the functions of the PECAM-1 gene and to determine which functional pathways of the PECA1 gene will involve in the human biological and pathological processes. (Fig.Ā 4C).
PECAM-1-targeted drug prediction analysis
The PECAM-1 targeted drug was detected with Targeted Medicinal Drugbank (https://go.drugbank.com/). A 3D structure of PECAM-1 was derived from PDB (https://www.rcsb.org/). The 3D structure of the small molecule of the predicated drugs was obtained from PubChem (https://pubchem.ncbi.nlm.nih.gov/). AutoDockTools 1.5.7 was employed to dock drug molecules that coordinate with PECAM-1 and predict their binding energy. Discovery Studio 4.5 was applied to visualize docking.
Statistical methods
Statistical Package for the Social Sciences 26.0 software (SPSS Inc., Chicago, IL, USA) was used for data analysis. Continuous variables were expressed as the meanā±āstandard deviation, while categorical variables were presented as percentages. Logistic regression was employed to analyze the independent risk factors for vascular injury. Pā<ā0.05 on both sides was considered to indicate a significant difference.
Results
Significant decrease of plasma PECAM-1 level in T2DMs
A total of 66 patients with T2DM (mean age 59.0ā±ā13.4 years, 45 males and 21 females) and 66 healthy individuals undergoing physical examinations (mean age 54.5ā±ā10.4 years, 42 males and 24 females) were enrolled in this study. Plasma samples and relevant clinical and biochemical data were collected, and plasma levels of PECAM-1 in the T2DM and healthy control groups were compared via ELISA. The results showed that compared with the healthy control group, T2DM patients had higher glycated hemoglobin (HbA1c) (9.3% vs. 5.3%), cholesterol (6.9 mmol/L vs. 4.9 mmol/L), triglyceride (1.2 mmol/L vs. 0.8 mmol/L), platelet count (Plt) (217.2āĆā109/L vs. 177.0āĆā109/L), and body mass index (BMI) (24.3Ā kg/m2 vs. 22.0Ā kg/m2), and fewer engaged in aerobic exercise (53.0% vs. 71.2%). Significantly, plasma PECAM-1 levels in T2DM patients were remarkably lower than those in healthy control group (18.8 ng/mL vs. 36.3 ng/mL) (TableĀ 1), suggesting that PECAM-1 may be implicated in the pathological mechanism of vascular injury in T2DM.
Decreased PECAM-1 is an independent risk factor for vascular injury in T2DM
T2DM patients were classified into a vascular injury group and a non - vascular injury group according to the presence of vascular injury. TableĀ 2 compares the PECAM-1 levels and relevant clinical and biochemical data between the two groups. The results show that the HbA1c level in the vascular injury group is significantly higher than that in the non - vascular injury group, more patients in the vascular injury group are smokers, and the PECAM-1 level in the vascular injury group is markedly lower (Pā<ā0.05). Univariate analysis indicates that elevated HbA1c, smoking habit, and decreased PECAM-1 level affect vascular injury in T2DM patients. Further multivariate regression analysis reveals that HbA1c and PECAM-1 are independent risk factors for vascular injury in T2DM patients. Meanwhile, collinearity analysis and visualization were performed for the variables in TableĀ 2. By calculating the Pearson correlation coefficient matrix of all variables, results showed that the absolute values of correlations between variables were all below 0.3, indicating no significant collinearity among variables (Fig.Ā 1A). Additionally, Variance Inflation Factor (VIF) was calculated as a supplementary analysis, with all variables demonstrating VIF valuesā<ā2.5, further confirming the robustness of the model (Fig.Ā 1B).
PECAM-1 transcription gene variation in T2DMs
Transcription factors play an important regulatory role in protein expression. In order to analyze the possible mechanism of PECAM-1 reduction in T2DM patients at the transcription level, we first detected the transcription factors involved in PECAM-1 transcription regulation. Based on the criteria of peak intensity signalā<ā500 and the predicted regulatory potential scoreā<ā1, the transcription factors involved in PECAM-1 regulation were analyzed through NetworkAnalyst. A total of 10 PECAM-1 regulating transcription factors, including responsive element binding protein 3 (CREB3), interferon Regulatory Factor 1 (IRF1), general transcription factor IIA subunit 2 (GTF2A2), nuclear receptor subfamily 2 group F member 6 (NR2F6), RELA proto-oncogene, NF-KB subunit (RELA), GATA zinc finger domain containing 1 (GATAD1), nuclear factor IA (NFIA ), REST corepressor 2 (RCOR2), cut like homeobox 1 (CUX1), and TEA domain transcription factor 3 (TEAD3), were found out (Fig.Ā 2A).
A mRNA dataset from the blood of T2DM patients (GSE26168; it includes the data from 9 T2DM patients and 8 normal controls) was obtained through the GEO public database. The results showed that comparing to the control group, the PECAM-1 transcription factors CREB3, GATAD1, and TEAD3 were significantly reduced (pā<ā0.01ā0.05) in T2DM, while CUX1 and RELA were significantly increased (pā<ā0.01) in T2DMs. There was no significant difference in other five transcription factors between the groups (Fig.Ā 2B). The results suggest that the variation of these transcription factors may be a regulatory mechanism for the decreased expression of PECAM-1 in T2DM plasma.
Differentially expressed genes in T2DM patients by GEO database analysis
Based on GSE26168 data analysis, a total of 100 significantly up-regulated genes (LogFC>1 ; p<0.05)(red) and 106 significantly down-regulated genes (LogFC<1 ; p<0.05) (blue) were obtained (Fig.Ā 3A). Among them, the top 20 significantly up-regulated genes (red) and the top 20 significantly down-regulated genes (green) were listed on a hot map (Fig.Ā 3B). OLFM4, CEACAM6 (CEA adhesion molecule 6), ITGB3 (integratin subunit beta 3), CEACAM8, and TMPRSS9 (transmembrane serine protease 9) genes which are related to vascular stability and permeability were significantly increased (LogFC>1; p<0.05). While, ARFIP2 (ADP ribosylation factor interacting protein 2) and TMCO5A (transmembrane and coiled-coil domains 5Ā A) were significantly reduced (LogFC<1; p<0.05). Noticeably, these significantly differential expressed proteins included those involving cellular adherent, cellular migration, cellular skeleton and membrane proteins, such as: CEACAM8 and 6, ITGB3, TMPRSS9, and ARFIP2. The results suggested that these abnormal expressed genes may involve the pathological mechanism of vascular injury in T2DMs.
Speculation of PECAM-1 bio-activities
The biological and pathological functional roles of PECAM-1 was analyzed using the STRING online network database. To easy understand the protein-protein interaction (PPI) related to PECAM-1, cytoscape was used to visualize the obtained PECAM-1-PPI proteins as a network diagram (Fig.Ā 4A). It revealed that kinase insert domain Receptor (KDR), cadherin 5 (CDH5), CDH17, selectin E (SELE), CD44, catenin beta 1 (CTNNB1), protein tyrosine phosphatase non-receptor type 1 (PTPN1), PTPN6, CD38, and CD177 tightly interact with PECAM-1, playing important roles in the regulation of vascular stability, endothelial development, and vascular permeability. The GO (Fig.Ā 4B) enrichment analysis was further performed using the ClusterProfiler package in R language. The results showed that PECAM-1 was involved in processes of plasma membrane adhesion, cell membrane adhesion via plasma membrane adhesion molecules, endometrial cell differentiation, endometrial development, peptidyl tyrosine modification, and cell cell junction organization, which closely relate to vascular permeability, endothelial development, endothelial cell functional information transmission, etc., including plasma-membrane adhesion, cell-cell adhesion via plasma-membrane adhesion molecules, endothelial cell differentiation, endothelium development, peptidylāāātyrosine phosphorylation, cellāāācell junction organization, basolateral plasma membrane, cadherin binding, etc. KEGG (Fig.Ā 4C) enrichment analysis also showed that PECAM-1 plays an important role in fluid shear stress and atherosclerosis, adherens junction, and cell adhesion molecules activities, all of them participate in vascular pathological and biological processes.
PECAM-1 targeted drug speculation
Given the above data, PECAM-1 may play an important role in vascular injury in T2DM. In order to explore the therapeutic medicine for vascular injuries in clinical treatment of T2DM, we employed Drugbank (https://go.drugbank.com/) analysis, a database that combines bioinformatics and chemical informatics resource analysis, to analyze and retrieve targeted drugs related to PECAM-1. The results showed that fostamatinib can dock with PECAM-1 molecule. Utilizing the small molecule 3D structure of fostamatinib (Fig.Ā 5A and B) obtained from PubChem (https://pubchem.ncbi.nlm.nih.gov/) and the PECAM-1 3D structure (Fig.Ā 5B) obtained from PDB (https://www.rcsb.org/), the molecular docking of them was verified using AutoDockTools 1.5.7, by which he predicted binding energy between them was āā7.1 (kcal/mol). FigureĀ 5 showed a 3D visualized images of PECAM-1-fostamatinib docking using Discovery Studio 4.5. This result indicated that fostamatinib could be a potential candidate drug for the treatment of vascular injuries in T2DMs.
Discussion
In this study, we initially detected the levels of plasma PECAM-1 in individuals with T2DM and those of healthy controls. We discovered that plasma PECAM-1 was significantly reduced in T2DM patients. Subsequently, we investigated the transcription factors involved in the regulation of PECAM-1 transcription using NetworkAnalyst. By analyzing T2DMsā blood mRNA data from the GEO database, we identified changes in certain PECAM-1 transcription factors and vascular regulatory factors in T2DM patients. Additionally, we explored the biological activity of PECAM-1 in vascular regulation using the STRING online network database. These findings and data indicated that PECAM-1 may have an important role in vascular injury in individuals with T2DM. Finally, we conducted a targeted drug matching analysis using the Drugbank database and found that fostamatinib may be a potential regulatory drug for PECAM-1, which could be applied in T2DMs with microvascular injuries.
Abnormal angiogenesis commonly occurs in almost all organs and tissues of either diabetic patients or experimental models. It includes the conditions such as diabetic retinopathy, nephropathy, microvascular injury to the axon of nerves, impaired wound healing, increased risk of heart disease, heart attacks, strokes, and narrowing of blood vessels. Recent research has shown that angiogenesis defects are also present in the lungs13, bone marrow14,15, and placenta16,17 of individuals with diabetes. Therefore, abnormal angiogenesis is a common feature of diabetic microvascular disease18,19,20. It is worth noting that abnormal angiogenesis is prevalent in diabetic complications, indeed, both excessive and defective angiogenesis can be observed in different tissues. Furthermore, diabetes is also associated with risks of coronary and peripheral artery diseases. Impaired angiogenesis can also affect the function of pancreatic islets and adipose tissue, worsening the damage caused by diabetes. The mechanisms of microvascular injury involve defective responses to hypoxia and pro-angiogenic factors, altered vasomotor tone, impaired shear stress sensing, inflammation, impaired nitric oxide bioavailability, lack of pro-angiogenic cells, and advanced glycation accumulation of end products, and pericyte loss18. Understanding the molecular factors that drive tissue-specific changes in diabetic microvascular injury is crucial for developing effective drugs, prevention strategies, and treatment methods for diabetic vascular injury.
PECAM-1 exists on the surface of platelets, monocytes, neutrophils and T cells, being an important protein in the inter-cellular connection of endothelial cells8,21.The adhesive properties of PECAM-1 largely depend on the α2,3-sialylated glycans participating PECAM-1/PECAM-1 formation. It regulates the vascular permeability barrier and leukocyte trafficking7. PECAM-1 is an important factor for vascular integrity as well7,9. Disruption of vascular integrity will lead to the accumulation of plasma, proteins and cells into the interstitial space, which is a signs of inflammation9.Thus, reducing PECAM-1 will induce a series of vascular injuries. Anti-PECAM-1 antibodies inhibit the tubular structures formation on Matrigel, reduce tumor-induced angiogenesis in mice. Tumor angiogenesis is impaired in PECAM-1-deficient mice7,8,22. Injection of PECAM-1 monoclonal antibody into mice resulted in fluid leakage into the liver and renal vasculature7. Inhibition of PECAM-1 reduced atherosclerotic lesions in mice23.In addition, PECAM-1 can interfere with the normal vascular function through inflammatory regulation. The plasma membrane fragment of PECAM-1 contains serines and tyrosines residues, both of which are present within the immunoreceptor tyrosine inhibitory motif (ITIM). The ITIM of PECAM-1 recruits the Src homology 2 containing protein tyrosine phosphatase2 when phosphorylated, thereby forming the PECAM-1/SHP-2 complex. This complex inhibits platelet activation, cytokine production and reactivity and stimulating prostacyclin production in the vessel wall, therefore prohibiting the activation of neutrophils, monocytes, and leukocytes7.Reduction of PECAM-1 will weaken its inhibitory effect on the activation of platelets and leukemia, leading to the activation of platelets and leukemia, inducing intravascular plaques, thrombus, and endothelial cell inflammation, resulting in vascular injury9,24. In this study, we found for the first time that plasma PECAM-1 level was significantly reduced in T2DM patients. According to the above mechanism, decreased PECAM-1 will cause vascular injury, including vascular instability, angiogenesis disorders, increased vascular permeability, local inflammation, thrombosis, etc.
Five regulatory factors, including CREB3, CUX1, GATAD1, RELA, and TEAD3, respectively, were found to be significant changed compared to those of the normal controls (p<0.05) from a T2DM blood mRNA sequencing data obtained from GEO public database (GSE26168) (Fig.Ā 2). In 2021, Gu et al. found that lead compound AG1296 had a vascular protective effect by activating factors such as CREB3 in a network pharmacology study25.Dong et al. demonstrated that sodium formononectin-3ā-sulphonate (Sul-F) promotes angiogenesis through regulation of cytokines including CREB3 in their study on a pharmalogical mechanism of Sul-F26. These data indicate that CREB3 exerts vascular protective and pro - angiogenic effects through certain mechanisms. GATAD1 gene mutation on the 7q21 chromosome arm leads to dilated cardiomyopathy27. Ma et al. reported that a significant decrease in the levels of GETAD1 and its 3 (prime) region DNA methylation in the placenta are positively correlated with the occurrence of preeclamptic placentas28. These results suggest that GATAD1 is an important cytokine in smooth muscle and vascular regulation. In a 9p21.3 Coronary Artery Disease study, Almontashiri et al. found that knocking down TEAD3 gene expression inhibited the expression of p16 that is a risk factor of Coronary Artery Disease, indicating that TEDAD3 is also closely related to vascular smooth muscle growth29.Based on our analysis, CREB3, GATAD1, and TEAD3 may exert their pathological effects in T2DM via the regulation of PECAM-1 gene transcription. A significant reduction of CREB3, GATAD1 and TEAD3 in T2DM patients is not conducive to vascular protection. CUX1 has a dual nature in tumor growth, either promoting or inhibiting tumor growth activities. The detailed mechanism is still unclear30.In a study on the activity of dipalmitoylphosphatidic acid (DPPA), Chen et al. reported that the anti-angiogenic activity of DPPA is created by suppression of CUX1 that reduce fibroblast growth factor 1 (FGF1) transcription31. It is suggested that CUX1 has a promoting effect on angiogenesis. RELA is functional domain of nuclear factor kappa B (NF-ĪŗB). In terms of vascular pathology, NF-ĪŗB can promote the occurrence and progression of arteriosclerosis in various ways and participates in cell proliferation in multiple ways32.Combining with our experimental data, increased CUX1 and RELA levels in T2DMs may promote abnormal angiogenesis and atherosclerosis through PECAM-1 transcription regulation.
A T2DM sequencing data (GSE26168) provided from the GEO database showed that CEACAM8, CEACAM6, ITGB3, and TMPRSS9 were significantly increased in T2DM blood, while ARFIP2 and TMCO5A were significantly reduced (Fig.Ā 3B). Carcinoembryonic antigen (CEA) is identified as a member of the CD66 differentiation cluster in immunology. These proteins include CD66b (CEACAM8), CD66c (CEACAM6), etc33.CEA is usually produced in the gastrointestinal tissue during fetal development, however it stops producing before birth and is elevated in many tumors, thus, being used as a tumor marker clinically. CEA can trigger endothelial cell behavior to promote angiogenesis, including adhesion, diffusion, proliferation, and migration in vitro, as well as tumor microvascularization in vivo34. Although we have not found any research report on TMPRSS9, however, its family member, TMPRSS4, is closely related to the vascular density of hepatocellular carcinoma (HCC), which can promote angiogenesis and HCC growth35, suggesting that TMPRSS9 may also have a similar activity. The increase of CEACAM6, CEACAM8, and TMPRSS9 in T2DMs may involve the induction of abnormal angiogenesis similar to cancer tissue. ITGB3 is the main integrin on platelets and crucial for platelet aggregation. It primarily involved in hemostasis and thrombosis36. The increase of ITGB3 in T2DMs is also consistent with the atherosclerotic lesions in T2DM patients, which should attract the attention of researchers to further explore the mechanism of ITGB3 in T2DM vascular injury. In a study of intracranial aneurysm (IA), Qin et al. found that the circ-ARFIP2 levels were significantly reduced in IA patients. Cellular experiments have revealed that circ-ARFIP2 enhances the proliferation, migration, and invasion ability of smooth muscle cells by regulating miR-338-3p37.Reduction of ARFIP2 in T2DM patients will affect the complete formation of blood vessels. Combining the angiogenic effects caused by elevated levels of CEACAM6, CEACAM8, and TMPRSS9, as well as the dysfunction of smooth muscle cells caused by decreased levels of ARFIP2, the formed novel blood vessels in T2DM patients might be dysfunctional blood vessels.
Interestingly, all proteins (Fig. 4A) associating to PECAM-1 obtained from the STRING online network database analysis are involved in angiogenesis and vascular function regulation. Among them, KDR is the receptor of VEGF (VEGFR-2), which mediates the regulation of endothelial cell function by VEGF38. CDH5 is the main endothelial adhesion molecule that controls cell connectivity and angiogenesis, which plays an important role in tumor vascular growth39. SEL and CD44 are involved in regulating inflammatory responses. Inflammation of the inner wall of blood vessels will increase vascular adhesion, leading to the accumulation of white blood cells, therefore, participating in the regulation of the pathogenesis of atherosclerosis40,41. CTNNB1 also takes part in inflammation and cellular oxidative responses, controlling various vascular regulatory factors42.CD38 participates in vascular remodeling via regulating vascular smooth muscle cells43. PTPN6 and PTPN11 are key regulatory components in cell cycle control, playing important roles in cell growth, proliferation, differentiation, and transformation control, including vascular endothelial cells44.CD177 also plays a positive role in tumor angiogenesis45. The above-mentioned and PECAM-1-involving vascular regulatory effects have also been validated in GO functional enrichment analysis. PECAM-1 participates in the following biological processes: plasma membrane adhesion, cell membrane adhesion via plasma membrane adhesion molecules, endometrial cell differentiation, endometrial development, peptidyl tyrosine modification, and cell cell junction organization (Fig. 4B). Furthermore, the functional enrichment analysis of KEGG also showed that PECAM-1 is involved in fluid shear stress and atmospheric clearance, adhesion junction, and cell adhesion molecules (Fig. 4C). Vascular endothelial injury and inflammation are central to the pathogenesis of vascular complications in T2DM, with PECAM-1 playing a critical role by regulating vascular barrier integrity, inflammatory cell migration, and endothelial mechanotransduction46. Previous studies have shown that PECAM-1 maintains endothelial barrier function by stabilizing adherens junctions, specifically through promoting the interaction between β-catenin and VE-cadherin, and its loss significantly enhances endothelial permeability to albumin9. During inflammation, PECAM-1 collaborates with CD99 and JAM (junctional adhesion molecule) to mediate leukocyte transendothelial migration: JAM facilitates leukocyte adhesion to endothelial cells via LFA-1 (lymphocyte function-associated antigen 1), PECAM-1 mediates subsequent extravasation, and CD99 completes the migration cascade47. Dysregulation of this process exacerbates vascular wall inflammation and promotes atherosclerotic plaque formation. In the context of T2DM, reduced PECAM-1 expression impairs endothelial cell response to shear stress, a key mechanical signal in vascular homeostasis, thereby disrupting vascular integrity48. Collectively, these findings indicate that PECAM-1 deficiency in T2DM contributes to vascular injury by compromising the endothelial barrier, promoting inflammatory cell infiltration, and inducing aberrant angiogenesis. Future intervention strategies targeting PECAM-1 may provide new directions for the treatment of vascular complications in T2DM.
Fostamatinib (commercial names as Tavalisse and Tavlese) is a tyrosine kinase inhibitor drug used clinically to treat chronic immune thrombocytopenia (ITP)49. Our study revealed that fostamatinib has a strong affinity for PECAM-1 (Fig.Ā 5). Platelets are the main source of PECAM-1 production, indicating the important role of PECAM-1 in platelet function regulation7. Based on the logical idea of using fostamatinib in clinical ITP treatment to enhance platelets50, combined with the special affinity of fostamatinib to PECAM-1 discovered in this study, it is suggested that the mechanism of fostamatinib in treating ITP to enhance platelets may be mediated via PECAM-1 regulation. This viewpoint needs to be validated by pharmacological research in future. Ingo et al. demonstrated that oral fostamatinib reduces atherosclerotic lesion size in low-density lipoprotein receptor-deficient mice, accompanied by decreased macrophage infiltration, increased smooth muscle cells and collagen content, and stabilized plaques51. Notably, fostamatinib has shown safety in clinical trials for rheumatoid arthritis52, a chronic inflammatory disease, which aligns with its proposed role in modulating inflammation and vascular remodeling in T2DM. Given that T2DM is often associated with platelet hyperreactivity and endothelial dysfunction, fostamatinibās dual inhibition of platelet activation and inflammatory pathways may confer pleiotropic benefits for microvascular protection. Combined with the important vascular regulation function of PECAM-1 described above, it is necessary to explore the possible therapeutic effect of fostamatinib in diabetes vascular injury, tumor, thrombocytopenia, atherosclerosis, and other vascular diseases in clinical practice. Meanwhile, this study also provides research ideas for the pharmacological mechanism of fostamatinib.
One limitation of this study is the lack of multivariate adjustment for common comorbidities (e.g., hypertension) and diabetes medications (e.g., insulin, statins, metformin). These factors may indirectly confound the results by influencing endothelial function, inflammatory responses, or PECAM-1 transcriptional regulation. Additionally, laboratory validation of key PECAM-1 signaling pathways was not performed, which limits the mechanistic understanding of our findings. Future studies should comprehensively collect data on comorbidities and medication use, coupled with experimental verification, to validate these results and further refine the mechanistic analysis.
Conclusion
The serum PECAM-1 levels in T2DM patients were significantly reduced compared with healthy controls. The decrease in transcription factors CREB3, TEAD3, and GATAD1, as well as the increase in RELA and CUX1, obtained from a T2DM blood test database, can explain the serum test findings. Further analysis of PECAM-1 functional enrichment revealed that PECAM-1 is involved in angiogenesis, vascular integrity, and endothelial functional regulation from multiple directions. The T2DM blood test data provided by GEO showed that the functional proteins CEACAM6/8, ITGB3, and TMPRSS9 related to PECAM-1 in the blood of T2DM patients were significantly increased, while ARFIP2 and TMCO5A were significantly reduced. These functional protein alterations may participate in the vascular injury mechanism of T2DM patients. Finally, network pharmacological analysis showed that fostamatinib is a drug with high affinity for PECAM-1. The treatment effect of fostamatinib for vascular injury complications in T2DM and other vascular injury diseases still requires further research and verification.
Data availability
The datasets generated during and/or analyzed during the current study are available from the corresponding author upon reasonable request.
References
Ceriello, A. & Prattichizzo, F. Variability of risk factors and diabetes complications. Cardiovasc. Diabetol. 20 (1), 101. https://doi.org/10.1186/s12933-021-01289-4 (2021).
Faselis, C. et al. Microvascular complications of type 2 diabetes mellitus. Curr. Vasc Pharmacol. 18 (2), 117ā124. https://doi.org/10.2174/1570161117666190502103733 (2020).
Gotsch, K., Klein, W. W., Leb, G., Pavek, P. & Passath, A. HƤmodynamische vergleichsuntersuchungen verschiedener Herzglykoside [Comparative hemodynamic studies on various heart glycosides]. Z. Kreislaufforsch. 61 (7), 577ā587 (1972).
Smyth, S. & Heron, A. Diabetes and obesity: the twin epidemics. Nat. Med. 12 (1), 75ā80. https://doi.org/10.1038/nm0106-75 (2006).
Liu, H., Wang, X., Gao, H., Yang, C. & Xie, C. Physiological and pathological characteristics of vascular endothelial injury in diabetes and the regulatory mechanism of autophagy. Front. Endocrinol. (Lausanne). 14, 1191426. https://doi.org/10.3389/fendo.2023.1191426 (2023).
Horton, W. B. & Barrett, E. J. Microvascular dysfunction in diabetes mellitus and cardiometabolic disease. Endocr. Rev. 42 (1), 29ā55. https://doi.org/10.1210/endrev/bnaa025 (2021).
Lertkiatmongkol, P., Liao, D., Mei, H., Hu, Y. & Newman, P. J. Endothelial functions of platelet/endothelial cell adhesion molecule-1 (CD31). Curr. Opin. Hematol. 23 (3), 253ā259. https://doi.org/10.1097/MOH.0000000000000239 (2016).
Liao, D., Mei, H., Hu, Y., Newman, D. K. & Newman, P. J. CRISPR-mediated deletion of the PECAM-1 cytoplasmic domain increases receptor lateral mobility and strengthens endothelial cell junctional integrity. Life Sci. 193, 186ā193. https://doi.org/10.1016/j.lfs.2017.11.002 (2018).
Privratsky, J. R. & Newman, P. J. PECAM-1: regulator of endothelial junctional integrity. Cell. Tissue Res. 355 (3), 607ā619. https://doi.org/10.1007/s00441-013-1779-3 (2014).
Alberti, K. G. & Zimmet, P. Z. Definition, diagnosis and classification of diabetes mellitus and its complications. Part 1: diagnosis and classification of diabetes mellitus provisional report of a WHO consultation. Diabet. Med. 15 (7), 539ā553 (1998).
Djaberi, R. et al. Non-invasive cardiac imaging techniques and vascular tools for the assessment of cardiovascular disease in type 2 diabetes mellitus. Diabetologia 51 (9), 1581ā1593. https://doi.org/10.1007/s00125-008-1062-4 (2008).
Lehmann, E. D., Riley, W. A., Clarkson, P. & Gosling, R. G. Non-invasive assessment of cardiovascular disease in diabetes mellitus. Lancet 350, S14āS9. https://doi.org/10.1016/s0140-6736(97)90023-4 (1997).
Fuso, L., Pitocco, D. & Antonelli-Incalzi, R. Diabetic lung, an underrated complication from restrictive functional pattern to pulmonary hypertension. Diabetes Metab. Res. Rev. 35 (6), e3159. https://doi.org/10.1002/dmrr.3159 (2019).
Oikawa, A. et al. Diabetes mellitus induces bone marrow microangiopathy. Arterioscler. Thromb. Vasc Biol. 30 (3), 498ā508. https://doi.org/10.1161/ATVBAHA.109.200154 (2010).
Spinetti, G. et al. Global remodeling of the vascular stem cell niche in bone marrow of diabetic patients: implication of the microRNA-155/FOXO3a signaling pathway. Circ. Res. 112 (3), 510ā522. https://doi.org/10.1161/CIRCRESAHA.112.300598 (2013).
Bhattacharjee, D. et al. Histopathological study with immunohistochemical expression of vascular endothelial growth factor in placentas of hyperglycemic and diabetic women. J. Lab. Phys. 9 (4), 227ā233. https://doi.org/10.4103/JLP.JLP_148_16 (2017).
Higgins, M., Felle, P., Mooney, E. E., Bannigan, J. & McAuliffe, F. M. Stereology of the placenta in type 1 and type 2 diabetes. Placenta 32 (8), 564ā569. https://doi.org/10.1016/j.placenta.2011.04.015 (2011).
Fadini, G. P., Albiero, M., Bonora, B. M. & Avogaro, A. Angiogenic abnormalities in diabetes mellitus: mechanistic and clinical aspects. J. Clin. Endocrinol. Metab. 104 (11), 5431ā5444. https://doi.org/10.1210/jc.2019-00980 (2019).
Gerstein, H. C. & Werstuck, G. H. Dysglycaemia, vasculopenia, and the chronic consequences of diabetes. Lancet Diabetes Endocrinol. 1 (1), 71ā78. https://doi.org/10.1016/S2213-8587(13)70025-1 (2013).
Stehouwer, C. D. A. Microvascular dysfunction and hyperglycemia: A vicious cycle with widespread consequences. Diabetes 67 (9), 1729ā1741. https://doi.org/10.2337/dbi17-0044 (2018).
Albelda, S. M., Muller, W. A., Buck, C. A. & Newman, P. J. Molecular and cellular properties of PECAM-1 (endoCAM/CD31): a novel vascular cell-cell adhesion molecule. J. Cell. Biol. 114 (5), 1059ā1068. https://doi.org/10.1083/jcb.114.5.1059 (1991).
Zhou, Z., Christofidou-Solomidou, M., Garlanda, C. & DeLisser, H. M. Antibody against murine PECAM-1 inhibits tumor angiogenesis in mice. Angiogenesis 3 (2), 181ā188. https://doi.org/10.1023/a:1009092107382 (1999).
Stevens, H. Y. et al. PECAM-1 is a critical mediator of atherosclerosis. Dis. Model. Mech. 1 (2ā3), 175ā181. https://doi.org/10.1242/dmm.000547 (2008). discussion 179.
Elias, C. G. 3rd et al. Ligation of CD31/PECAM-1 modulates the function of lymphocytes, monocytes and neutrophils. Eur. J. Immunol. 28(6), 1948-58 (1998).
Gu, M. et al. iPSC-endothelial cell phenotypic drug screening and in Silico analyses identify tyrphostin-AG1296 for pulmonary arterial hypertension. Sci. Transl. Med. 13 (592), eaba6480. https://doi.org/10.1126/scitranslmed.aba6480 (2021).
Dong, Z. et al. Sulphonated Formononetin induces angiogenesis through vascular endothelial growth factor/camp response Element-Binding protein/early growth response 3/vascular cell adhesion molecule 1 and Wnt/β-Catenin signaling pathway. Pharmacology 101 (1ā2), 76ā85. https://doi.org/10.1159/000480662 (2018).
Yang, J., Shah, S., Olson, T. M. & Xu, X. Modeling GATAD1-Associated dilated cardiomyopathy in adult zebrafish. J. Cardiovasc. Dev. Dis. 3 (1), 6. https://doi.org/10.3390/jcdd3010006 (2016).
Ma, X., Li, J., Brost, B., Cheng, W. & Jiang, S. W. Decreased expression and DNA methylation levels of GATAD1 in preeclamptic placentas. Cell. Signal. 26 (5), 959ā967. https://doi.org/10.1016/j.cellsig.2014.01.013 (2014).
Almontashiri, N. A. et al. 9p21.3 coronary artery disease risk variants disrupt TEAD transcription factor-Dependent transforming growth factor β regulation of p16 expression in human aortic smooth muscle cells. Circulation 132 (21), 1969ā1978. https://doi.org/10.1161/CIRCULATIONAHA.114.015023 (2015).
Ramdzan, Z. M. & Nepveu, A. CUX1, a haploinsufficient tumour suppressor gene overexpressed in advanced cancers. Nat. Rev. Cancer. 14 (10), 673ā682. https://doi.org/10.1038/nrc3805 (2014).
Chen, J. et al. Dipalmitoylphosphatidic acid inhibits breast cancer growth by suppressing angiogenesis via Inhibition of the CUX1/FGF1/HGF signalling pathway. J. Cell. Mol. Med. 22 (10), 4760ā4770. https://doi.org/10.1111/jcmm.13727 (2018).
De Martin, R., Hoeth, M., Hofer-Warbinek, R. & Schmid, J. A. The transcription factor NF-kappa B and the regulation of vascular cell function. Arterioscler. Thromb. Vasc. Biol. 20 (11), E83āE88. https://doi.org/10.1161/01.atv.20.11.e83 (2000).
Duffy, M. J. Carcinoembryonic antigen as a marker for colorectal cancer: is it clinically useful? Clin. Chem. 47 (4), 624ā630 (2001).
Bramswig, K. H. et al. Soluble carcinoembryonic antigen activates endothelial cells and tumor angiogenesis. Cancer Res. 73 (22), 6584ā6596. https://doi.org/10.1158/0008-5472.CAN-13-0123 (2013).
Dong, Z. R. et al. TMPRSS4 drives angiogenesis in hepatocellular carcinoma by promoting HB-EGF expression and proteolytic cleavage. Hepatology 72 (3), 923ā939. https://doi.org/10.1002/hep.31076 (2020).
Ma, Y. Q., Qin, J. & Plow, E. F. Platelet integrin alpha(IIb)beta(3): activation mechanisms. J. Thromb. Haemost. 5 (7), 1345ā1352. https://doi.org/10.1111/j.1538-7836.2007.02537.x (2007).
Qin, K., Tian, G., Zhou, D., Chen, G. & Circular RNA circ-ARFIP2 regulates proliferation, migration and invasion in human vascular smooth muscle cells via miR-338-3p-dependent modulation of KDR. Metab. Brain Dis. 36 (6), 1277ā1288. https://doi.org/10.1007/s11011-021-00726-3 (2021).
Holmes, K., Roberts, O. L., Thomas, A. M. & Cross, M. J. Vascular endothelial growth factor receptor-2: structure, function, intracellular signalling and therapeutic Inhibition. Cell. Signal. 19 (10), 2003ā2012. https://doi.org/10.1016/j.cellsig.2007.05.013 (2007).
Nan, W., He, Y., Wang, S. & Zhang, Y. Molecular mechanism of VE-cadherin in regulating endothelial cell behaviour during angiogenesis. Front. Physiol. 14, 1234104. https://doi.org/10.3389/fphys.2023.1234104 (2023).
Chase, S. D., Magnani, J. L. & Simon, S. I. E-selectin ligands as mechanosensitive receptors on neutrophils in health and disease. Ann. Biomed. Eng. 40 (4), 849ā859. https://doi.org/10.1007/s10439-011-0507-y (2012).
Krolikoski, M., Monslow, J. & PurĆ©, E. The CD44-HA axis and inflammation in atherosclerosis: A Temporal perspective. Matrix Biol. 78ā79, 201ā218. https://doi.org/10.1016/j.matbio.2018.05.007 (2019).
Vallée, A. & Lecarpentier, Y. Crosstalk between peroxisome Proliferator-Activated receptor gamma and the canonical WNT/β-Catenin pathway in chronic inflammation and oxidative stress during carcinogenesis. Front. Immunol. 9, 745. https://doi.org/10.3389/fimmu.2018.00745 (2018).
Gan, L. et al. CD38 deficiency alleviates Ang II-induced vascular remodeling by inhibiting small extracellular vesicle-mediated vascular smooth muscle cell senescence in mice. Signal. Transduct. Target. Ther. 6 (1), 223. https://doi.org/10.1038/s41392-021-00625-0 (2021).
Vestweber, D. Vascular endothelial protein tyrosine phosphatase regulates endothelial function. Physiol. (Bethesda). 36 (2), 84ā93. https://doi.org/10.1152/physiol.00026.2020 (2021).
Jiang, J., Chen, Y., Zhang, M., Zhou, H. & Wu, H. Relationship between CD177 and the vasculogenic mimicry, clinicopathological parameters, and prognosis of epithelial ovarian cancer. Ann. Palliat. Med. 9 (6), 3985ā3992. https://doi.org/10.21037/apm-20-1825 (2020).
Kong, P. et al. Inflammation and atherosclerosis: signaling pathways and therapeutic intervention. Signal. Transduct. Target. Ther. 7 (1), 131. https://doi.org/10.1038/s41392-022-00955-7 (2022).
Privratsky, J. R., Newman, D. K. & Newman, P. J. PECAM-1: conflicts of interest in inflammation. Life Sci. 87 (3ā4), 69ā82. https://doi.org/10.1016/j.lfs.2010.06.001 (2010).
Xie, X. et al. Low shear stress induces endothelial cell apoptosis and monocyte adhesion by upregulating PECAM-1 expression. Mol. Med. Rep. 21 (6), 2580ā2588. https://doi.org/10.3892/mmr.2020.11060 (2020).
Fostamatinib [M]. Drugs and Lactation Database (LactMedĀ®). Bethesda. (2006).
Paik, J. & Fostamatinib A Review in Chronic Immune Thrombocytopenia. Drugs 81(8), 935ā943 https://doi.org/10.1007/s40265-021-01540-y (2021).
Hilgendorf, I. et al. The oral spleen tyrosine kinase inhibitor fostamatinib attenuates inflammation and atherogenesis in low-density lipoprotein receptor-deficient mice. Arterioscler. Thromb. Vasc. Biol. 31 (9), 1991ā1999. https://doi.org/10.1161/ATVBAHA.111.230847 (2011).
Tanaka, Y., Millson, D., Iwata, S. & Nakayamada, S. Safety and efficacy of fostamatinib in rheumatoid arthritis patients with an inadequate response to methotrexate in phase II OSKIRA-ASIA-1 and OSKIRA-ASIA-1X study. Rheumatol. (Oxford). 60 (6), 2884ā2895. https://doi.org/10.1093/rheumatology/keaa732 (2021).
Acknowledgements
We express our gratitude to all of the participants in the study, including the patients and their families.
Author information
Authors and Affiliations
Contributions
JJ-X and XM-C designed the study. HZ-C, H-S, and X-C collected samples and clinical data of the T2DM patients and the healthy controls. HZ-C performed experiments. JJ-X and XM-C conduct statistical analysis. JJ-X wrote the manuscript. All authors have reviewed and approved the final manuscript, confirming its content and agreeing to be accountable for all aspects of the work.
Corresponding author
Ethics declarations
Competing interests
The authors declare no competing interests.
Additional information
Publisherās note
Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Rights and permissions
Open Access This article is licensed under a Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International License, which permits any non-commercial use, sharing, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if you modified the licensed material. You do not have permission under this licence to share adapted material derived from this article or parts of it. The images or other third party material in this article are included in the articleās Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the articleās Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by-nc-nd/4.0/.
About this article
Cite this article
Xu, JJ., Cai, HZ., Sun, H. et al. Decreased PECAM-1 May be a potential pathological factor for vascular injury in T2DM patients. Sci Rep 15, 22001 (2025). https://doi.org/10.1038/s41598-025-04915-3
Received:
Accepted:
Published:
DOI: https://doi.org/10.1038/s41598-025-04915-3