Open Access

Role of transcription factor FOXM1 in diabetes and its complications (Review)

  • Authors:
    • Baoqing Zhao
    • Mengxi Li
    • Yanting Su
    • Shigang Shan
    • Wenbin Qian
    • Dan Zhu
    • Xiufen Liu
    • Zhenwang Zhang
  • View Affiliations

  • Published online on: September 5, 2023     https://doi.org/10.3892/ijmm.2023.5304
  • Article Number: 101
  • Copyright: © Zhao et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Diabetes mellitus is a chronic metabolic disease commonly associated with complications such as cardiovascular disease, nephropathy and neuropathy, the incidence of which is increasing yearly. Transcription factor forkhead box M1 (FOXM1) serves an important role in development of diabetes and its complications. The present study aimed to review the association between FOXM1 with pathogenesis of diabetes and its complications. FOXM1 may be involved in development and progression of diabetes and its complications by regulating cell biological processes such as cell cycle, DNA damage repair, cell differentiation and epithelial‑mesenchymal transition. FOXM1 is involved in regulation of insulin secretion and insulin resistance, and FOXM1 affects insulin secretion by regulating expression of insulin‑related genes and signaling pathways; FOXM1 is involved in the inflammatory response in diabetes, and FOXM1 can regulate key genes associated with inflammatory response and immune cells, which in turn affects occurrence and development of the inflammatory response; finally, FOXM1 is involved in the regulation of diabetic complications such as cardiovascular disease, nephropathy and neuropathy. In summary, the transcription factor FOXM1 serves an important role in development of diabetes and its complications. Future studies should explore the mechanism of FOXM1 in diabetes and find new targets of FOXM1 as a potential treatment for diabetes and its complications.

1. Introduction

Diabetes mellitus (DM) is a common metabolic disease, and the number of people aged 20-79 with diabetes is expected to increase to 642 million by 2040 (1). Patients with diabetes often exhibit complications, such as cardiovascular disease, neuropathy and retinopathy, which have impact patient life and health (2,3). Therefore, it is important to study the pathogenesis and treatment of diabetes and its complications (4-6). The transcription factor forkhead box M1 (FOXM1) is widely present in a variety of cells and serves an important regulatory role in biological processes such as cell cycle, DNA damage repair and cell differentiation (7,8). FOXM1 also serves an important role in development of diabetes and its complications (9-31). FOXM1 is involved in regulating the cell cycle, promoting cell proliferation, maintaining normal cellular differentiation and DNA damage repair (11,12,32). In addition, FOXM1 may be involved in regulating onset and progression of diabetic complications, such as cardiovascular disease and nephropathy (13,33). The present study aimed to review the role of FOXM1 in diabetic complications in terms of its expression, regulatory mechanism and function, and provide new ideas and approaches for the prevention and treatment of diabetes. Studies have shown that FOXM1 is abnormally expressed in diabetic patients, especially in patients with diabetic foot ulcer (DFU) (9,10,15,31), cardiovascular disease (11,12), diabetic nephropathy (DN) (14), neuropathy (25,26) and erectile dysfunction (Fig. 1) (30). The aforementioned studies suggest that FOXM1 may be involved in regulation of the development of diabetes and its complications. Studies have shown that multiple signaling pathways and molecules regulate the expression and activity of FOXM1, such as YAP1/Akt/glycogen synthase kinase-3 (GSK3β)/FOXM1 (11), VEGF/FOXM1 (30), pituitary adenylate cyclase-activating polypeptide (PACAP)/FOXM1 (25,34), vasoactive intestinal polypeptide (VIP)/FOXM1 (25,35), IL-6/STAT3/FOXM1 (26,27), PI3K/Akt/FOXM1 (36), Ras-ERK/FOXM1 and JNK/p38MAPK/FOXM1 (7). Therefore, FOXM1 may be a key node in the regulation of these signaling pathways and molecules in diabetes and its complications. FOXM1 can regulate the expression of various genes, such as the cell cycle regulatory genes polo-like Kinase 1 (PLK1) and Centromere protein A (CENP-A) (18,29), DNA damage repair genes (Aurora kinase B, Baculoviral IAP Repeat-Containing Protein 5, BUB1 mitotic checkpoint serine/threonine kinase B, centromere protein E, Signal STAT3) (9) and apoptosis-associated genes growth Arrest-specific Transcripts (GAS5), TATA-Box Binding Protein Associated Factor 15 (TAF15) and stromal cell derived factor 4 (SDF4) (31). In addition, some studies have found that FOXM1 may also regulate DM and its complications through the function of immune cells (including T and B cells, monocytes, macrophages, and dendritic cells) (9,10,37). Thus, FOXM1 may be involved in development of diabetes and its complications by regulating the expression of these genes.

In the present study, PubMed database was searched for 'FOXM1 and diabetes mellitus and its complications' between 2000 and 2023 (pubmed.ncbi.nlm.nih.gov/?term=FOXM1+and+diabetes+mellitus+and+its+complications&filter=dates.2000%2F1%2F1-2023). Results were mainly published in the past five years. The present study reviews the role and function of FOXM1 in diabetes and its complications through gene expression and gene regulation. It was hypothesized that FOXM1 is involved in regulating insulin (INS) secretion and resistance, the inflammatory response and development and progression of complications in diabetes. Studies have confirmed that FOXM1 is a key regulator of the development of diabetes and its complications (9-31).

2. FOXM1 in regulation of INS secretion and resistance

FOXM1 is a transcription factor, in islet cells FOXM1 is involved in the regulation of INS secretion (38,39) (Fig. 2). Studies have shown that in islet cells, FOXM1 regulates the expression of INS secretion-related genes, or is regulated to promote INS synthesis and secretion, including INS, type 1 glucose transporter (GLUT1), GLUT2, INS-like growth factor-I (IGF-I), leptin and adiponectin (38-40). Saavedra-García et al (41) confirmed that the FOXO3-FOXM1 axis regulates fatty acid metabolism. Studies have shown that glucokinase-mediated glucose metabolism promotes adaptive β-cell proliferation-induced INS synthesis and secretion via activation of the FOXM1/PLK1/CENP-A pathway (18,25,29,36,37). Acetylcholine, PACAP and VIP promote adaptive β-cell proliferation via upregulation of FOXM1 (25,42,43). STAT3/FOXM1/GLUT1 pathway signaling regulates aerobic glycolysis and upregulation of GLUT expression promotes glucose-stimulated INS secretion in human and mouse β-cells (44,45). Glucagon-like peptide-1 (GLP-1) receptor agonists and dipeptidyl peptidase-4 (DPP-4) inhibitors potentially compensate for dysregulated β-cell glucose metabolism (18).

In addition, FOXM1 is involved in regulation of INS resistance (IR). The causes of IR in obesity and type 2 DM (T2DM) are not limited to impaired INS signaling but also involve complex interactions of multiple metabolic pathways. Metabolites are regulated by modulating components of INS signaling pathways such as JAK2/STAT3, IKK/NF-κB, JNK/p38MAPK and PI3K/AKT signaling pathways (46-50). Zarrouki et al (51) found that plasma FOXM1 levels are increased and positively correlated with IR. FOXM1 mediates multiple IR-associated JAK2/STAT3, NF-κB and PI3K/Akt pathways. FOXM1 expression could be regulated through the JAK2/STAT3 pathway (52); upregulation of FOXM1 increases NF-κB expression (53). FOXO3 and FOXM1 forkhead box transcription factors downstream of PI3K/Akt and JNK/p38MAPK signaling cascades act downstream of PI3K/Akt and JNK/p38MAPK signaling cascades and are key for cell proliferation, differentiation, cell survival, senescence, DNA damage repair and cell cycle control (7).

FOXM1 can also influence the development and progression of IR through pathways such as regulation of fatty acid metabolism and inflammatory responses. Elevated free fatty acid (FFA)-induced lipotoxicity may play an important role in the pathogenesis of β-cell IR (33,54,55). Mangiferin improves IR in HepG2 and C2C12 cells by increasing glucose consumption and promoting serum FFA oxidation via the PPARα pathway (56). Oxidative phosphorylation (OXPHOS) is linked to increased levels of reactive oxygen species (ROS), which are key signaling molecules; production of excess ROS can have deleterious effects, ultimately leading to cell death. By linking FOXM1-dependent peroxiredoxin-3 (Prx3) expression and fatty acid oxidation-mediated NADPH regeneration, Cancer stem cells (CSCs) with increased levels of OXPHOS maintain low ROS levels. FOXM1-dependent ROS regulation is an intracellular resistance mechanism in cancer dry-like cells (57). Nutritional overload triggers an uncontrolled inflammatory response, leading to chronic low-grade inflammation that promotes IR (58). In particular, overexpression of FOXM1 inhibits the inflammatory response following myocardial infarction, thereby protecting the heart. Conversely, inhibition of FOXM1 activity abolishes the cardioprotective effect (59).

Taken together, FOXM1 serves an important role in the regulation of INS secretion and resistance and may be a potential target for the treatment of diabetes.

3. FOXM1 in the inflammatory response in diabetes

FOXM1 is involved in regulating the inflammatory response associated with diabetes. FOXM1 is involved in pathogenesis of diabetes by regulating the expression of inflammation-associated mediators (IL-1, IL-6, TNF-α) and cellular chemokines (monocyte Chemoattractant Protein-1, C-X3-C motif chemokine ligand 1) are expressed and involved in the pathogenesis of diabetes (60-63). In addition, FOXM1 also regulates certain inflammation-associated signaling pathways, such as Wnt/β-catenin (64), JAK/STAT (65), NF-κB (66), SMAD3 (67) and P38MAPK (7) (Fig. 3). FOXM1 deubiquitination could be induced through the Wnt/β-catenin signaling pathway, stabilizing the structure and expression of FOXM1, thereby transporting β-catenin into the nucleus and promoting the transcription of genes related to the biological behavior of inflammation (68). FOXM1 activates the Wnt/β-catenin via transcriptional regulation of multiple Wnt expression signaling pathways and promotes renal fibrosis (64). Silencing of FOXM1 prevents the effects of apoptosis, inflammation, NF-κB and JAK/STAT signaling pathway activation. Moreover, the expression of FOXM1 is positively correlated with the expression of p38MAPK and NF-κB and induces expression of inflammatory factors and amplification of the inflammatory response cascade via the p38MAPK/NF-κB signaling pathway (65). It has been proposed that FOXM1 can directly interact with Smad3 protein to promote Smad3 nuclear retention and bind to β-catenin promoter sequence to promote fibrosis (69). microRNA-4429 targeting FOXM1 can decrease expression of SMAD3 and impede cell proliferation, migration, invasion and epithelial-mesenchymal transition. High expression of FOXM1 increases secretion of T helper and 2 cytokines and promotes inflammatory responses mediated by PI3K/AKT/GSK-33 and p38MAPK signaling pathways. These findings suggest that FOXM1 serves an important regulatory role in the inflammatory response. Therefore, FOXM1 may be a potential target for diabetes therapy.

In DFUs, major transcriptional networks lead to decreased neutrophil and macrophage recruitment and a poorly controlled inflammatory response. The functions of transcription factors FOXM1 and STAT3 are required to activate and promote survival of immune cells, yet expression of transcription factors FOXM1 and STAT3 are suppressed in DFUs. Furthermore, inhibition of FOXM1 (streptozotocin-induced and Leprdb mutant mice-db/db) in a diabetic mouse model results in delayed diabetic wound healing and reduced neutrophil and macrophage recruitment in vivo. This shows that a disordered and ineffective inflammatory response is a key factor in the pathogenesis of DFUs, which is facilitated by FOXM1-mediated relaxation of neutrophil and macrophage recruitment, revealing a potential therapeutic strategy (9).

FOXM1 modulates the function of immune cells in diabetic patients, thereby influencing the extent and duration of the inflammatory response (9,37). Thus, FOXM1 may be a potential target for the treatment of diabetes and associated immune diseases. Future studies should explore the mechanism of FOXM1 action in immune cells in diabetic patients and develop FOXM1 inhibitors as novel drugs for the treatment of diabetes and associated immune diseases.

4. FOXM1 in regulating the development and progression of diabetic complications

FOXM1 serves an important role in the complications of DM. Persistent hyperglycemia, hyperinsulinemia and subsequent oxidative stress lead to diabetic complications, primarily classified as microvascular (nephropathy, retinopathy and neuropathy) and macrovascular (cardiomyopathy) complications (70). Studies have shown that FOXM1 is abnormally expressed in diabetic patients with nephropathy, retinopathy, cardiovascular disease, DFU and tumors (9-12,14,71). FOXM1 is involved in the development and progression of complications of diabetes via multiple pathways, including regulation of cell proliferation, apoptosis, inflammatory response, oxidative stress and angiogenesis. In DN renal tissue, FOXM1 is lowly expressed. FOXM1 overexpression improves renal function in mice, decreases pathological changes and increases the expression of the podocyte marker nephrin in renal tissue (14). In diabetic retinopathy (DR), FOXO1 is aberrantly expressed and the FOXM1/FOXO1 axis is involved in regulating development and progression of DR (72,73). In diabetic cardiovascular complications, FOXM1 is overexpressed in myocardial tissue in a hyperglycemic state, leading to increased cardiomyocyte hypertrophy and fibrotic response (11). Maternally imprinted gene (MEG3) promotes degradation of FOXM1 protein by promoting FOXM1 ubiquitination, thereby decreasing VEGF expression and ultimately regulating endothelial differentiation of bone marrow mesenchymal stem cells (BM-MSCs). MEG3/FOXM1 controls differentiation of BM-MSCs to endothelial cells (ECs) (30). FOXM1 regulates ROS levels in neutrophils and inhibition of FOXM1 leads to an increase in ROS, resulting in neutrophil extracellular trap (NET) formation, leading to tissue damage and impaired healing (10). Therefore, FOXM1 serves an important role in the complications of diabetes and may be a new target for the prevention and treatment of diabetic complications.

FOXM1 is involved in DN

DN is a renal microvascular complication caused by DM, characterized by proteinuria and progressive renal injury, and is a key contributor to end-stage renal disease (74-76). The most typical pathological changes in renal biopsies of patients with DN are glomerular lesions, primarily including diffuse and nodular thylakoid expansion and glomerular basement membrane thickening (77).

FOXM1-activated sirtuin (SIRT)4 inhibits NF-κB signaling and NLRP3 inflammatory vesicles to attenuate renal injury and podocyte apoptosis in diabetic kidney disease (14). The overexpression of FOXM1 improves renal function and decreases pathological changes in mice, and it increases the expression of the foot cell marker nephrin in kidney tissue. In in vitro experiments, FOXM1 increases the viability and decreases pyroptosis of high Glucose-treated MPC5 cells and increases the expression of the podocyte marker nephrin, while decreasing the expression of NLRP3 inflammatory vesicles and cleaved caspase 1 associated with scorch death (14). In conclusion, FOXM1 alleviates renal injury and podocyte pyroptosis in patients with DN by transcriptionally activating SIRT4 and inhibiting the NF-κB signaling pathway and NLRP3 inflammatory vesicles (Fig. 4).

Activation of the Wnt/β-catenin pathway serves a key role in promoting renal fibrosis. The transcription factor FOXM1 is significantly increased in the kidney of patients with obstructive kidney and fibrosis (64). FoxM1 is mainly distributed in the renal tubular epithelial cells. In renal tubular epithelial cells, overexpression of FoxM1 promotes the expression of eight Wnts, whereas knockdown of FoxM1 inhibits angiotensin II)-induced expression of multiple Wnts, including Wnt1, Wnt2b and Wnt3, and FoxM1 regulates the transcription of Wnt1, Wnt2b, and Wnt3. Inhibition of FoxM1 downregulates expression of Wnts, ultimately leading to the reduction of renal fibrosis (64). These findings suggest that FoxM1 may be a key switch in activating the β-catenin pathway and renal fibrosis (Fig. 4). Therefore, FoxM1 may be a potential therapeutic target for the renal fibrosis (64).

Acute kidney injury (AKI) is characterized by sudden loss of renal function due to tubular epithelial damage. Renal tubular regeneration is essential to prevent progression of chronic kidney disease (78). Renal FoxM1 expression is increased in mice following renal ischemia/reperfusion (I/R)-induced AKI. Thiostrepton targeting of FoxM1 decreases FoxM1-regulated pro-proliferative factors and cell proliferation in vitro, as well as tubular regeneration in mice following AKI, indicating FoxM1 is important for tubular regeneration after AKI (78). FOXM1 inhibits diabetic kidney injury and promotes tissue repair by regulating SIRT4 expression; FOXM1 activates β-catenin, Cyclin D1 and c-myc, and inhibits p21 and p27 expression to induce tubular regeneration after AKI. In addition, FOXM1, induced early in injury, is required for epithelial cell proliferation in vitro, and is dependent on epidermal growth factor receptor (EGFR) stimulation (79).

FOXM1 regulates multiple signaling pathways and gene expression, including TGF-β, Wnt/β-catenin and NF-κB (64,80,81), as well as cell cycle, apoptosis and oxidative stress (82,83). Thus, it affects the proliferation, apoptosis and differentiation of cells, including glomerular and tubular cells (81). Therefore, FOXM1 may be a new target for treatment of DN. Researchers are exploring the signaling pathways and mechanisms of action regulated by FOXM1, as well as potential FOXM1 application in DN diagnosis and treatment (64,80-83).

FOXM1 may be involved in regulation of DR

Oxidative stress and inflammation are key causative factors for DR, which is the most common ocular complication of DM and the leading cause of visual impairment in working-age people worldwide. FOXM1 regulates cell cycle, proliferation, apoptosis and metabolism by affecting the expression of microvascular cell inflammation (IL-1β, IL-6, TNF-α and NF-κB) and apoptosis genes (caspase-3) and may be involved in the regulation of DR. INS blocking leads to elevated blood sugar levels and may lead to excess triggering of FOXM1, ultimately increasing the production of several apoptotic and inflammatory factors such as TNF-α and NF-κB, as well as ROS, which may also lead to DR (16,20,23,66).

FOXM1 can regulate the expression of retinopathy-related genes such as VEGF and TGF-β, thereby affecting the occurrence and progression of DR (84-87). Therefore, FOXM1 may serve an important role in the occurrence and development of DR. To the best of our knowledge, however, there are no studies on the regulatory role of FOXM1 in DR.

FOXM1 is involved in diabetic neuropathy

Diabetic neuropathy is a common complication that affects function of the nervous system, including sensory, motor and autonomic systems (42). By blocking the hepatic branch of the vagus nerve and inducing deletion or overexpression of hepatocyte-specific FOXM1, it has been shown that vagal signaling is involved in the inhibition or rapid activation of the hepatocyte FOXM1 pathway and affects hepatocyte proliferation following partial hepatectomy. Vagotomy increases postoperative mortality and replenishment of hepatic FOXM1 prevents this, which is a key mechanism to improve survival following liver injury. Notably, macrophages act as mediators of vagal signaling and acute activation of the FOXM1 pathway in hepatocytes (27). Acetylcholine secreted by the vagus nerve and IL-6 secreted by macrophages are involved in the molecular mechanism of vagus-macrophage-hepatocyte transduction by which STAT3 is phosphorylated via the IL-6 signaling pathway and phosphorylated STAT3 binds to the FOXM1 gene promoter, and the activated FOXM1 gene promotes hepatocyte regeneration (27,88). Vagal signaling directly activates the FOXM1 pathway in pancreatic β cells, thus promoting compensatory proliferation (42,89). Unlike islet cells, which are abundantly distributed, vagus innervation is scarce in the liver and only visible around the portal vein area. Thus, vagal signaling-mediated IL-6 production in hepatic macrophages upregulates hepatocyte FOXM1, leading to liver regeneration and ensuring survival. This complex multi-step mechanism of neuronal, immune and parenchymal cells allows emergency regenerative signals to be amplified and propagated through the organ, thus promoting rapid liver regeneration and ensuring systemic survival following severe liver injury (27,88,89). Studies suggest that FOXM1 may be involved in development and progression of diabetic neuropathy through mechanisms such as regulation of neuronal apoptosis, oxidative stress and inflammation (27,42,88,89). However, more studies are needed to determine the exact role and mechanisms of FOXM1 in diabetic neuropathy.

FOXM1 is involved in diabetic cardiovascular disease

FOXM1 is involved in the development and progression of diabetic cardiovascular disease. During diabetes, sustained hyperglycemic stress in cardiac primary contractile cells and cardiomyocytes leads to increased apoptosis, which leads to cardiomyocyte hypertrophy and fibrosis (90). Cardiac hypertrophy and fibrosis lead to structural and functional abnormalities (such as arrhythmia, heart failure and atrial fibrillation) that contribute to risk of heart failure. FOXM1 is overexpressed in myocardial tissue during hyperglycemia, leading to increased cardiomyocyte hypertrophic and fibrotic responses (11) (Fig. 5). Under hyperglycemic stress, YAP1 in cardiomyocytes leads to elevated AKT phosphorylation by activating YAP1, and elevated AKT mediates loss of inhibitory regulation of FOXM1 by GSK3β inactivation. Thus, upregulated YAP1 leads to abnormal FOXM1 accumulation in cardiomyocytes. This elevated FOXM1 promotes pathological remodeling of cardiomyocytes, leading to cardiomyocyte hypertrophy and fibrosis. FOXM1 serves an important role in cardiovascular pathology (11). FOXM1 may promote the development of diabetic cardiovascular disease. Endothelial and smooth muscle cells mediate vascular remodeling via FoxM1 signaling interactions (91); FOXM1 promotes vascular endothelial cell and cardiomyocyte proliferation and neonatal heart regeneration (92). FOXM1 may also be involved in the development of diabetic cardiovascular disease. FOXM1 can regulate cardiomyocyte proliferation and thus affect cardiomyocyte survival and function. During zebrafish heart regeneration following injury, Foxm1 is required in cardiomyocyte proliferation through transcriptional regulation of cell cycle genes (32). FOXM1 regulates biological processes such as cardiac fibrosis and inflammatory responses, accelerating the deterioration of cardiac function (11). Previous studies have shown that FOXM1 serves a key role in inflammation and inflammatory cell recruitment (10,14,15). FOXM1 regulates the expression of pro-inflammatory cytokines and chemokines, including IL-6, inducible nitric oxide synthase (INOS), Chemokine C-C-Motif Receptor 2 (CCR2) and CX3C chemokine receptor1 (CX3CR1) (93-96).

ERK acts upstream of the FOXM1 transcription factor, which is a key downstream effector of the ERK pathway involved in the control of cycle progression and cell proliferation and activates it via phosphorylation and nuclear translocation. FOXM1 expression is induced by tissue damage and is decreased in senescent cells (97,98). Therefore, FOXM1 is associated with the pathogenesis of atherosclerosis and may be involved in diabetic atherosclerosis (13).

Hyperglycemia, a feature of diabetes, can induce vascular complications by increasing endothelial cell apoptosis and limiting proliferation. The potential role of FoxM1 in high-glucose-induced EC injury, where FoxM1 protects EC from high-glucose-induced growth arrest and apoptosis by inhibiting ROS induced by regulation of the Akt and ERK pathways, may be a novel therapeutic target to treat EC dysfunction (12). Therefore, the role of FOXM1 in diabetic cardiovascular disease may serve as a targets and strategies for the prevention and treatment of diabetic cardiovascular disease.

FOXM1 is involved in DFU

DFU is a serious complication caused by DM, associated with decreased quality of life and high mortality, with deterioration leading to foot amputation (99-101). FOXM1 controls oxidative stress by inducing the expression of ROS (such as Superoxide dismutase (SOD2, glutathione peroxidase 4, Serine/threonine protein phosphatase, Glycerol phosphate dehydrogenase, Serine hydroxymethyltransferase 2 and Methylene tetrahydrofolate dehydrogenase 2) (57,102). The wound environment of DFU is accompanied by prolonged inflammation, leading to impaired wound healing. FOXM1 is lowly expressed in DFU rat wound tissue and its ability to enhance DFU macrophage 2 (M2) polarization and trauma healing is enhanced; silencing FOXM1 reverses promotion of M2 polarization-induced human dermal fibroblast proliferation and migration (Fig. 6). Thus, targeting the transcription factor FOXM1 may provide a therapeutic target for promoting DFU wound healing (15).

In a diabetic mouse model of skin injury, inhibition of FOXM1 results in delayed wound closure and decreased recruitment of neutrophils and macrophages (9). FOXM1 is a regulator of neutrophil response during acute skin and oral mucosal wound healing in human DFU (9).

The FOXM1 signaling pathway regulates NET formation during diabetic wound healing. FOXM1 is a regulator of NET formation by modulating ROS levels to promote neutrophil immune responses during wound healing (103,104). Furthermore, triggering receptor Expressed on Myeloid cells 1 (TREM1) is associated with FOXM1 during DFU wound healing, and TREM1 activation increases the recruitment of FOXM1-positive neutrophils and enhances diabetic wound healing (10,105). FOXM1 is downregulated in DFU, resulting in decreased neutrophil response, suggesting that TREM1/FOXM1 is a key regulator of diabetic wound healing. TREM1 promotes wound healing by regulating Interlukin-1β (IL1B), IL6, Cyclin Dependent Kinase 1 (CDK1), Amphiregulin (AREG), SOD2 and C-X-C motif chemokine 8 (CXCL8) which promote FOXM1 gene activation (10). TREM1/FOXM1 promotes the recruitment of neutrophils, reverses the effects of diabetes, and promotes wound healing in vivo, suggesting that the FOXM1 pathway is a novel regulator of NET formation during diabetic wound healing and revealing a new therapeutic strategy to promote DFU healing (10).

The expression of FOXM1, GAS5 and SDF4 is decreased in the skin tissue of patients with DFU (31). High glucose stimulation induces endoplasmic reticulum stress and apoptosis, which inhibits angiogenesis in Human umbilical vein endothelial cells (HUVECs), whereas FOXM1 overexpression alleviates inhibition of angiogenesis. FOXM1 regulates GAS5 expression and knockdown of GAS5 reverses the effects of FOXM1 overexpression. FOXM1 inhibits endoplasmic reticulum stress and apoptosis and promotes angiogenesis by mediating the GAS5/TAF15/SDF4 axis generation, providing a novel molecular mechanism for the treatment of DFU (31).

FOXM1 is involved in diabetic patients with tumors

Epidemiological studies have shown that DM and hyperglycemia are not only risk factors (106), but also poor prognostic indicators for numerous types of cancer (such as kidney, oesophageal, colorectal, breast and bladder cancer, and leukaemia) (107,108). A previous study found that high glucose-induced KKU-213A and KKU-213B cells exhibit high FOXM1 expression in a dose-dependent manner (22).

Obesity and inflammation are associated with increased risk of hepatocellular carcinoma, which is the leading cause of cancer-associated deaths worldwide. Tissue metalloproteinase inhibitor 3 (Timp3) deficiency during hepatocarcinogenesis in obesity is associated with decreased FoxM1 transcriptional activity via the H19/microRNA (miR)-675/p53 pathway (109). Timp3 ablation leads to cell cycle perturbation by suppressing FoxM1 transcriptional activity through the H19/miR-675/p53 pathway (109). Investigators have designed antitumor and antidiabetic drugs targeting FOXM1 (17,110).

Studies have identified that FOXM1 serves a key role in INS secretion and islet cell proliferation and thus FOXM1 inhibitors may be useful in improving IR and glycemic cont rol (18,25,29,36,37,111,112). FOXM1 inhibitors include thiostrepton and siomycin A, which inhibit FOXM1 transcriptional activity in cellular and animal models (17,110). However, further studies are needed to validate the efficacy and safety of inhibitors for use in clinical therapy.

5. Conclusion

DM and its complications are a notable public health problem worldwide and impact quality of life and survival of patients. FOXM1 is an important transcription factor that has been shown to serve a key role in the development and progression of DM and its complications (9,10,15,31). The present study reviewed the role and function of FOXM1 in diabetes and its complications. FOXM1 serves an important role in INS secretion and resistance (112). FOXM1 can promote INS synthesis and secretion, while inhibiting IR. In addition, FOXM1 affects the balance of glucose metabolism by regulating expression of genes associated with glucose metabolism. Second, the role of FOXM1 in diabetic complications has also received attention. Studies have shown that FOXM1 can be involved in occurrence and development of diabetes-associated complications such as nephropathy, neuropathy, and retinopathy (9-31). FOXM1 affects the occurrence and development of diabetic complications by regulating biological processes such as cell proliferation, apoptosis, and oxidative stress. Further research is needed to determine the specific regulatory mechanism of FOXM1, interaction with other transcription factors and the role of FOXM1 in different types and stages of diabetes. Future studies should investigate the role and function of FOXM1 in diabetes and its complications to provide novel approaches for the prevention and treatment of diabetes.

Despite need for effective treatments to cure diabetic complications such as DFU, no new Food and Drug Administration-approved therapy has been effective since 1998. The pathogenesis of diabetic complications involves intrinsic factors, such as neuropathy, vasculopathy, ischemia, infection, fibrosis and immune dysfunction. In conclusion, further development of drugs for the treatment of diabetes and its complications is required.

Availability of data and materials

Not applicable.

Authors' contributions

ZZ, DZ and XL designed the study and wrote the manuscript. BZ, ML, YS, SS and WQ performed the literature review. Data authentication is not applicable. All authors have read and approved the final manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Acknowledgments

Not applicable.

Funding

The present study was supported by Hubei Provincial Department of Education 'Hundred Schools and Hundred Counties' (grant no. BXLBX0806), Foundation of Hubei Educational Committee (grant nos. Q20202803), Foundation of Hubei University of Science and Technology Science 'Special Project on Diabetes and Angiopathy' (grant nos. BK202028, BK202010 and 2020TNB10/2022TNB11), Foundation of Hubei University of Science and Technology 'Double Hundred Project' (grant no. 2022HKSB01), Foundation of Innovation Team of Hubei University of Science and Technology (grant no. 2023T13) and Natural Science Foundation of Hubei Province (grant nos. 2021DFE025 and 2023AFB1027).

References

1 

Ogurtsova K, da Rocha Fernandes JD, Huang Y, Linnenkamp U, Guariguata L, Cho NH, Cavan D, Shaw JE and Makaroff LE: IDF Diabetes Atlas: Global estimates for the prevalence of diabetes for 2015-2040. Diabetes Res Clin Pract. 128:40–50. 2017. View Article : Google Scholar : PubMed/NCBI

2 

Tomic D, Shaw JE and Magliano DJ: The burden and risks of emerging complications of diabetes mellitus. Nat Rev Endocrinol. 18:525–539. 2022. View Article : Google Scholar : PubMed/NCBI

3 

Huang HK, Liu PP, Lin SM, Hsu JY, Yeh JI, Lai EC, Peng CC, Munir KM, Loh CH and Tu YK: Diabetes-Related complications and mortality in patients with atrial fibrillation receiving different oral anticoagulants: A nationwide analysis. Ann Intern Med. 175:490–498. 2022. View Article : Google Scholar : PubMed/NCBI

4 

Zhu S, Li J and Zhao X: Comparative risk of new-onset hyperkalemia for antihypertensive drugs in patients with diabetic nephropathy: A Bayesian network meta-analysis. Int J Clin Pract. 75:e139402021. View Article : Google Scholar

5 

Kong L, Zhao H, Fan J, Wang Q, Li J, Bai J and Mao J: Predictors of frailty among Chinese community-dwelling older adults with type 2 diabetes: A cross-sectional survey. BMJ Open. 11:e0415782021. View Article : Google Scholar : PubMed/NCBI

6 

Chen Y, Wu G and Xu M: The effect of L-thyroxine substitution on oxidative stress in early-stage diabetic nephropathy patients with subclinical hypothyroidism: A randomized double-blind and placebo-controlled study. Int Urol Nephrol. 50:97–103. 2018. View Article : Google Scholar

7 

Yao S, Fan LY and Lam EW: The FOXO3-FOXM1 axis: A key cancer drug target and a modulator of cancer drug resistance. Semin Cancer Biol. 50:77–89. 2018. View Article : Google Scholar

8 

Zhang Z, Liu W, Bao X, Sun T, Wang J, Li M and Liu C: USP39 facilitates breast cancer cell proliferation through stabilization of FOXM1. Am J Cancer Res. 12:3644–3661. 2022.PubMed/NCBI

9 

Sawaya AP, Stone RC, Brooks SR, Pastar I, Jozic I, Hasneen K, O'Neill K, Mehdizadeh S, Head CR, Strbo N, et al: Deregulated immune cell recruitment orchestrated by FOXM1 impairs human diabetic wound healing. Nat Commun. 11:46782020. View Article : Google Scholar : PubMed/NCBI

10 

Sawaya AP, Stone RC, Mehdizadeh S, Pastar I, Worrell S, Balukoff NC, Kaplan MJ, Tomic-Canic M and Morasso MI: FOXM1 network in association with TREM1 suppression regulates NET formation in diabetic foot ulcers. EMBO Rep. 23:e545582022. View Article : Google Scholar : PubMed/NCBI

11 

Mondal A, Das S, Samanta J, Chakraborty S and Sengupta A: YAP1 induces hyperglycemic stress-mediated cardiac hypertrophy and fibrosis in an AKT-FOXM1 dependent signaling pathway. Arch Biochem Biophys. 722:1091982022. View Article : Google Scholar : PubMed/NCBI

12 

Cao J, Jiang X and Peng X: Forkhead box M1 inhibits endothelial cell apoptosis and cell-cycle arrest through ROS generation. Int J Clin Exp Pathol. 11:4899–4907. 2018.PubMed/NCBI

13 

Hamledari H, Sajjadi SF, Alikhah A, Boroumand MA and Behmanesh M: ASGR1 but not FOXM1 expression decreases in the peripheral blood mononuclear cells of diabetic atherosclerotic patients. J Diabetes Complications. 33:539–546. 2019. View Article : Google Scholar : PubMed/NCBI

14 

Xu X, Zhang L, Hua F, Zhang C, Zhang C, Mi X, Qin N, Wang J, Zhu A, Qin Z and Zhou F: FOXM1-activated SIRT4 inhibits NF-κB signaling and NLRP3 inflammasome to alleviate kidney injury and podocyte pyroptosis in diabetic nephropathy. Exp Cell Res. 408:1128632021. View Article : Google Scholar

15 

Yang Y, Zhang B, Yang Y, Peng B and Ye R: FOXM1 accelerates wound healing in diabetic foot ulcer by inducing M2 macrophage polarization through a mechanism involving SEMA3C/NRP2/Hedgehog signaling. Diabetes Res Clin Pract. 184:1091212022. View Article : Google Scholar

16 

Gerst F, Kemter E, Lorza-Gil E, Kaiser G, Fritz AK, Nano R, Piemonti L, Gauder M, Dahl A, Nadalin S, et al: The hepatokine fetuin-A disrupts functional maturation of pancreatic beta cells. Diabetologia. 64:1358–1374. 2021. View Article : Google Scholar : PubMed/NCBI

17 

Tabatabaei Dakhili SA, Perez DJ, Gopal K, Haque M, Ussher JR, Kashfi K and Velázquez-Martínez CA: SP1-independent inhibition of FOXM1 by modified thiazolidinediones. Eur J Med Chem. 209:1129022021. View Article : Google Scholar

18 

Shirakawa J and Terauchi Y: Newer perspective on the coupling between glucose-mediated signaling and β-cell functionality. Endocr J. 67:1–8. 2020. View Article : Google Scholar

19 

Ma J, Xing B, Cao Y, He X, Bennett KE, Tong C, An C, Hojnacki T, Feng Z, Deng S, et al: Menin-regulated Pbk controls high fat diet-induced compensatory beta cell proliferation. EMBO Mol Med. 13:e135242021. View Article : Google Scholar : PubMed/NCBI

20 

Kohata M, Imai J, Izumi T, Yamamoto J, Kawana Y, Endo A, Sugawara H, Seike J, Kubo H, Komamura H, et al: Roles of FoxM1-driven basal β-cell proliferation in maintenance of β-cell mass and glucose tolerance during adulthood. J Diabetes Investig. 13:1666–1676. 2022. View Article : Google Scholar : PubMed/NCBI

21 

Chen HY, Chen DT, Chiang YY, Lin SY and Lee CN: The correlation of forkhead box protein M1 (FOXM1) with gestational diabetes mellitus in maternal peripheral blood and neonatal umbilical cord blood. Taiwan J Obstet Gynecol. 61:652–656. 2022. View Article : Google Scholar : PubMed/NCBI

22 

Detarya M, Thaenkaew S, Seubwai W, Indramanee S, Phoomak C, Saengboonmee C, Wongkham S and Wongkham C: High glucose upregulates FOXM1 expression via EGFR/STAT3 dependent activation to promote progression of cholangiocarcinoma. Life Sci. 271:1191142021. View Article : Google Scholar : PubMed/NCBI

23 

Peng G, Mosleh E, Yuhas A, Katada K, Cherry C and Golson ML: FOXM1 acts sexually dimorphically to regulate functional β-cell mass. bioRxiv. Jan 12–2023.Epub ahead of print.

24 

Zhong L, Zhao Z, Hu Q, Li Y, Zhao W, Li C, Xu Y, Rong R, Zhang J, Zhang Z, et al: Identification of maturity-onset diabetes of the young caused by mutation in FOXM1 via whole-exome sequencing in Northern China. Front Endocrinol (Lausanne). 11:–534362. 2021. View Article : Google Scholar

25 

Imai J: Regulation of compensatory β-cell proliferation by inter-organ networks from the liver to pancreatic β-cells. Endocr J. 65:677–684. 2018. View Article : Google Scholar : PubMed/NCBI

26 

Imai J: Regulation of adaptive cell proliferation by vagal nerve signals for maintenance of whole-body homeostasis: Potential therapeutic target for insulin-deficient diabetes. Tohoku J Exp Med. 254:245–252. 2021. View Article : Google Scholar : PubMed/NCBI

27 

Izumi T, Imai J, Yamamoto J, Kawana Y, Endo A, Sugawara H, Kohata M, Asai Y, Takahashi K, Kodama S, et al: Vagus-macrophage-hepatocyte link promotes post-injury liver regeneration and whole-body survival through hepatic FoxM1 activation. Nat Commun. 9:53002018. View Article : Google Scholar : PubMed/NCBI

28 

Chen M, Zhao S, Guo WH, Zhu YP, Pan L, Xie ZW, Sun WL and Jiang JT: Maternal exposure to Di-n-butyl phthalate (DBP) aggravate gestational diabetes mellitus via FoxM1 suppression by pSTAT1 signalling. Ecotoxicol Environ Saf. 205:1111542020. View Article : Google Scholar : PubMed/NCBI

29 

Shirakawa J, Tajima K, Okuyama T, Kyohara M, Togashi Y, De Jesus DF, Basile G, Kin T, Shapiro AMJ, Kulkarni RN and Terauchi Y: Luseogliflozin increases beta cell proliferation through humoral factors that activate an insulin receptor- and IGF-1 receptor-independent pathway. Diabetologia. 63:577–587. 2020. View Article : Google Scholar : PubMed/NCBI

30 

Sun X, Luo LH, Feng L and Li DS: Down-regulation of lncRNA MEG3 promotes endothelial differentiation of bone marrow derived mesenchymal stem cells in repairing erectile dysfunction. Life Sci. 208:246–252. 2018. View Article : Google Scholar : PubMed/NCBI

31 

Peng W, Zhu T, Xiang G, Ding T, Zhao J, Xiong D, Zhong Y and Zhang Y: Identification of signalling downstream of the transcription factor forkhead box protein M1 that protects against endoplasmic reticulum stress in a diabetic foot ulcer model. Diabet Med. 40:e150512023. View Article : Google Scholar : PubMed/NCBI

32 

Zuppo DA, Missinato MA, Santana-Santos L, Li G, Benos PV and Tsang M: Foxm1 regulates cardiomyocyte proliferation in adult zebrafish after cardiac injury. Development. 150:dev2011632023. View Article : Google Scholar : PubMed/NCBI

33 

Zhang Z, Li M, Sun T, Zhang Z and Liu C: FOXM1: Functional roles of FOXM1 in Non-Malignant diseases. Biomolecules. 13:8572023. View Article : Google Scholar : PubMed/NCBI

34 

Filipsson K, Sundler F, Hannibal J and Ahren B: PACAP and PACAP receptors in insulin producing tissues: Localization and effects. Regul Pept. 74:167–175. 1998. View Article : Google Scholar : PubMed/NCBI

35 

Ahren B: Islet G protein-coupled receptors as potential targets for treatment of type 2 diabetes. Nat Rev Drug Discov. 8:369–385. 2009. View Article : Google Scholar : PubMed/NCBI

36 

Zhang L, Jiang B, Zhu N, Tao M, Jun Y, Chen X, Wang Q and Luo C: Mitotic checkpoint kinase Mps1/TTK predicts prognosis of colon cancer patients and regulates tumor proliferation and differentiation via PKCα/ERK1/2 and PI3K/Akt pathway. Med Oncol. 37:52019. View Article : Google Scholar

37 

Zheng J, Bu X, Wei X, Ma X and Zhao P: The role of FoxM1 in immune cells. Clin Exp Med. Mar 13–2023.Epub ahead of print.

38 

Zhang H, Ackermann AM, Gusarova GA, Lowe D, Feng X, Kopsombut UG, Costa RH and Gannon M: The FoxM1 transcription factor is required to maintain pancreatic beta-cell mass. Mol Endocrinol. 20:1853–1866. 2006. View Article : Google Scholar : PubMed/NCBI

39 

Mahmoodzadeh Sagheb M, Azarpira N, Mokhtary M, Hosseini SE and Yaghobi R: The effects of Leptin and Adiponectin on Pdx1, Foxm1, and PPARү Transcription in rat islets of langerhans. Hepat Mon. 13:e90552013.

40 

Liu MN, Zhang L, Dong XY, Liu M, Cheng G, Zhang XL, He F and Wang GQ: Effects of Akkermansia muciniphila on the proliferation, apoptosis and insulin secretion of rat islet cell tumor cells. Sichuan Da Xue Xue Bao Yi Xue Ban. 51:13–17. 2020.In Chinese. PubMed/NCBI

41 

Saavedra-García P, Nichols K, Mahmud Z, Fan LY and Lam EW: Unravelling the role of fatty acid metabolism in cancer through the FOXO3-FOXM1 axis. Mol Cell Endocrinol. 462(Pt B): 82–92. 2018. View Article : Google Scholar

42 

Yamamoto J, Imai J, Izumi T, Takahashi H, Kawana Y, Takahashi K, Kodama S, Kaneko K, Gao J, Uno K, et al: Neuronal signals regulate obesity induced β-cell proliferation by FoxM1 dependent mechanism. Nat Commun. 8:19302017. View Article : Google Scholar

43 

Imai J and Katagiri H: Regulation of systemic metabolism by the autonomic nervous system consisting of afferent and efferent innervation. Int Immunol. 34:67–79. 2022. View Article : Google Scholar

44 

Shang R, Wang M, Dai B, Du J, Wang J, Liu Z, Qu S, Yang X, Liu J, Xia C, et al: Long noncoding RNA SLC2A1-AS1 regulates aerobic glycolysis and progression in hepatocellular carcinoma via inhibiting the STAT3/FOXM1/GLUT1 pathway. Mol Oncol. 14:1381–1396. 2020. View Article : Google Scholar : PubMed/NCBI

45 

Yang Y, Cai Z, Pan Z, Liu F, Li D, Ji Y, Zhong J, Luo H, Hu S, Song L, et al: Rheb1 promotes glucose-stimulated insulin secretion in human and mouse β-cells by upregulating GLUT expression. Metabolism. 123:1548632021. View Article : Google Scholar

46 

Heo YJ, Choi SE, Jeon JY, Han SJ, Kim DJ, Kang Y, Lee KW and Kim HJ: Visfatin induces inflammation and insulin resistance via the NF-κB and STAT3 signaling pathways in hepatocytes. J Diabetes Res. 2019:40216232019. View Article : Google Scholar

47 

Alaaeldin R, Abdel-Rahman IAM, Hassan HA, Youssef N, Allam AE, Abdelwahab SF, Zhao QL and Fathy M: Carpachromene ameliorates insulin resistance in HepG2 cells via modulating IR/IRS1/PI3k/Akt/GSK3/FoxO1 pathway. Molecules. 26:76292021. View Article : Google Scholar : PubMed/NCBI

48 

Gao W, Du X, Lei L, Wang H, Zhang M, Wang Z and Li X, Liu G and Li X: NEFA-induced ROS impaired insulin signalling through the JNK and p38MAPK pathways in non-alcoholic steatohepatitis. J Cell Mol Med. 22:3408–3422. 2018. View Article : Google Scholar : PubMed/NCBI

49 

Shrestha S, Kumar Singh V, Kumar Sarkar S, Shanmugasundaram B, Jeevaratnam K and Chandra Koner B: Effect of sub-toxic exposure to Malathion on glucose uptake and insulin signaling in L6 myoblast derived myotubes. Drug Chem Toxicol. 43:663–670. 2020. View Article : Google Scholar

50 

Saltiel AR: Insulin signaling in health and disease. J Clin Invest. 131:e1422412021. View Article : Google Scholar : PubMed/NCBI

51 

Zarrouki B, Benterki I, Fontés G, Peyot ML, Seda O, Prentki M and Poitout V: Epidermal growth factor receptor signaling promotes pancreatic β-cell proliferation in response to nutrient excess in rats through mTOR and FOXM1. Diabetes. 63:982–993. 2014. View Article : Google Scholar :

52 

Zhao C, Chen HY, Zhao F, Feng HJ and Su JP: Acylglycerol kinase promotes paclitaxel resistance in nasopharyngeal carcinoma cells by regulating FOXM1 via the JAK2/STAT3 pathway. Cytokine. 148:1555952021. View Article : Google Scholar : PubMed/NCBI

53 

Li Y, Lu L, Tu J, Zhang J, Xiong T, Fan W, Wang J, Li M, Chen Y, Steggerda J, et al: Reciprocal regulation between forkhead box M1/NF-κB and methionine adenosyltransferase 1A drives liver cancer. Hepatology. 72:1682–1700. 2020. View Article : Google Scholar : PubMed/NCBI

54 

Bodis K and Roden M: Energy metabolism of white adipose tissue and insulin resistance in humans. Eur J Clin Invest. 48:e130172018. View Article : Google Scholar : PubMed/NCBI

55 

Zheng S, Chen N, Kang X, Hu Y and Shi S: Irisin alleviates FFA induced β-cell insulin resistance and inflammatory response through activating PI3K/AKT/FOXO1 signaling pathway. Endocrine. 75:740–751. 2022. View Article : Google Scholar

56 

Zhang Q, Kong X, Yuan H, Guan H, Li Y and Niu Y: Mangiferin improved palmitate-induced-insulin resistance by promoting free fatty acid metabolism in HepG2 and C2C12 cells via PPARα: Mangiferin improved insulin resistance. J Diabetes Res. 2019:20526752019. View Article : Google Scholar

57 

Choi HJ, Jhe YL, Kim J, Lim JY, Lee JE, Shin MK and Cheong JH: FoxM1-dependent and fatty acid oxidation-mediated ROS modulation is a cell-intrinsic drug resistance mechanism in cancer stem-like cells. Redox Biol. 36:1015892020. View Article : Google Scholar : PubMed/NCBI

58 

Longo M, Zatterale F, Naderi J, Parrillo L, Formisano P, Raciti GA, Beguinot F and Miele C: Adipose tissue dysfunction as determinant of obesity-associated metabolic complications. Int J Mol Sci. 20:23582019. View Article : Google Scholar : PubMed/NCBI

59 

Tian S, Lei I, Gao W, Liu L, Guo Y, Creech J, Herron TJ, Xian S, Ma PX, Eugene Chen Y, et al: HDAC inhibitor valproic acid protects heart function through Foxm1 pathway after acute myocardial infarction. EBioMedicine. 39:83–94. 2019. View Article : Google Scholar :

60 

Tang SCW and Yiu WH: Innate immunity in diabetic kidney disease. Nat Rev Nephrol. 16:206–222. 2020. View Article : Google Scholar : PubMed/NCBI

61 

Vaidya VS, Niewczas MA, Ficociello LH, Johnson AC, Collings FB, Warram JH, Krolewski AS and Bonventre JV: Regression of microalbuminuria in type 1 diabetes is associated with lower levels of urinary tubular injury biomarkers, kidney injury molecule-1, and N-acetyl-β-D-glucosaminidase. Kidney Int. 79:464–470. 2011. View Article : Google Scholar

62 

Alicic RZ, Johnson EJ and Tuttle KR: Inflammatory mechanisms as new biomarkers and therapeutic targets for diabetic kidney disease. Adv Chronic Kidney Dis. 25:181–191. 2018. View Article : Google Scholar : PubMed/NCBI

63 

Nastase MV, Zeng-Brouwers J, Beckmann J, Tredup C, Christen U, Radeke HH, Wygrecka M and Schaefer L: Biglycan, a novel trigger of Th1 and Th17 cell recruitment into the kidney. Matrix Biol. 68-69:293–317. 2018. View Article : Google Scholar

64 

Xie H, Miao N, Xu D, Zhou Z, Ni J, Yin F, Wang Y, Cheng Q, Chen P, Li J, et al: FoxM1 promotes Wnt/β-catenin pathway activation and renal fibrosis via transcriptionally regulating multi-Wnts expressions. J Cell Mol Med. 25:1958–1971. 2021. View Article : Google Scholar : PubMed/NCBI

65 

Zhu-Ge D, Yang YP and Jiang ZJ: Knockdown CRNDE alleviates LPS-induced inflammation injury via FOXM1 in WI-38 cells. Biomed Pharmacother. 103:1678–1687. 2018. View Article : Google Scholar : PubMed/NCBI

66 

Zhou M, Shi J, Lan S and Gong X: FOXM1 regulates the proliferation, apoptosis and inflammatory response of keratinocytes through the NF-κB signaling pathway. Hum Exp Toxicol. 40:1130–1140. 2021. View Article : Google Scholar : PubMed/NCBI

67 

Song S, Zhang R, Cao W, Fang G, Yu Y, Wan Y, Wang C, Li Y and Wang Q: Foxm1 is a critical driver of TGF-β-induced EndMT in endothelial cells through Smad2/3 and binds to the Snail promoter. J Cell Physiol. 234:9052–9064. 2019. View Article : Google Scholar

68 

Chen Y, Li Y, Xue J, Gong A, Yu G, Zhou A, Lin K, Zhang S, Zhang N, Gottardi CJ and Huang S: Wnt-induced deubiquitination FoxM1 ensures nucleus β-catenin transactivation. EMBO J. 35:668–684. 2016. View Article : Google Scholar : PubMed/NCBI

69 

Xie H, Gao YM, Zhang YC, Jia MW, Peng F, Meng QH and Wang YC: Low let-7d exosomes from pulmonary vascular endothelial cells drive lung pericyte fibrosis through the TGFβRI/FoxM1/Smad/β-catenin pathway. J Cell Mol Med. 24:13913–13926. 2020. View Article : Google Scholar : PubMed/NCBI

70 

Jalgaonkar MP, Parmar UM, Kulkarni YA and Oza MJ: SIRT1-FOXOs activity regulates diabetic complications. Pharmacol Res. 175:1060142022. View Article : Google Scholar

71 

Inoue Y, Moriwaki K, Ueda Y, Takeuchi T, Higuchi K and Asahi M: Elevated O-GlcNAcylation stabilizes FOXM1 by its reduced degradation through GSK-3β inactivation in a human gastric carcinoma cell line, MKN45 cells. Biochem Biophys Res Commun. 495:1681–1687. 2018. View Article : Google Scholar

72 

Behl T, Wadhwa M, Sehgal A, Singh S, Sharma N, Bhatia S, Al-Harrasi A, Aleya L and Bungau S: Mechanistic insights into the role of FOXO in diabetic retinopathy. Am J Transl Res. 14:3584–3602. 2022.PubMed/NCBI

73 

Sengupta A, Kalinichenko VV and Yutzey KE: FoxO1 and FoxM1 transcription factors have antagonistic functions in neonatal cardiomyocyte cell-cycle withdrawal and IGF1 gene regulation. Circ Res. 112:267–277. 2013. View Article : Google Scholar

74 

Ioannou K: Diabetic nephropathy: Is it always there? Assumptions, weaknesses and pitfalls in the diagnosis. Hormones (Athens). 16:351–361. 2017.

75 

Sanajou D, Ghorbani Haghjo A, Argani H and Aslani S: AGE-RAGE axis blockade in diabetic nephropathy: Current status and future directions. Eur J Pharmacol. 833:158–164. 2018. View Article : Google Scholar : PubMed/NCBI

76 

Tziomalos K and Athyros VG: Diabetic Nephropathy: New risk factors and improvements in diagnosis. Rev Diabet Stud. 12:110–118. 2015. View Article : Google Scholar : PubMed/NCBI

77 

Qi C, Mao X, Zhang Z and Wu H: Classification and differential diagnosis of diabetic nephropathy. J Diabetes Res. 2017:86371382017. View Article : Google Scholar : PubMed/NCBI

78 

Sinha S, Dwivedi N, Woodgett J, Tao S, Howard C, Fields TA, Jamadar A and Rao R: Glycogen synthase kinase-3β inhibits tubular regeneration in acute kidney injury by a FoxM1-dependent mechanism. FASEB J. 34:13597–13608. 2020. View Article : Google Scholar : PubMed/NCBI

79 

Chang-Panesso M, Kadyrov FF, Lalli M, Wu H, Ikeda S, Kefaloyianni E, Abdelmageed MM, Herrlich A, Kobayashi A and Humphreys BD: FOXM1 drives proximal tubule proliferation during repair from acute ischemic kidney injury. J Clin Invest. 129:5501–5517. 2019. View Article : Google Scholar : PubMed/NCBI

80 

Wang Y, Zhou Q, Tang R, Huang Y and He T: FoxM1 inhibition ameliorates renal interstitial fibrosis by decreasing extracellular matrix and epithelial-mesenchymal transition. J Pharmacol Sci. 143:281–289. 2020. View Article : Google Scholar : PubMed/NCBI

81 

Yu W, Wang G, Li LX, Zhang H, Gui X, Zhou JX, Calvet JP and Li X: Transcription factor FoxM1 promotes cyst growth in PKD1 mutant ADPKD. Hum Mol Genet. 32:1114–1126. 2023. View Article : Google Scholar

82 

Halasi M and Gartel AL: Targeting FOXM1 in cancer. Biochem Pharmacol. 85:644–652. 2013. View Article : Google Scholar

83 

Liao GB, Li XZ, Zeng S, Liu C, Yang SM, Yang L, Hu CJ and Bai JY: Regulation of the master regulator FOXM1 in cancer. Cell Commun Signal. 16:572018. View Article : Google Scholar : PubMed/NCBI

84 

Zhu X, Yu M, Wang K, Zou W and Zhu L: FoxM1 affects adhesive, migratory, and invasive abilities of human retinoblastoma Y-79 cells by targeting matrix metalloproteinase 2. Acta Biochim Biophys Sin (Shanghai). 52:294–301. 2020. View Article : Google Scholar : PubMed/NCBI

85 

Zhu X, Xue L, Yao Y, Wang K, Tan C, Zhuang M, Zhou F and Zhu L: The FoxM1-ABCC4 axis mediates carboplatin resistance in human retinoblastoma Y-79 cells. Acta Biochim Biophys Sin (Shanghai). 50:914–920. 2018. View Article : Google Scholar : PubMed/NCBI

86 

Wang RT, Miao RC, Zhang X, Yang GH, Mu YP, Zhang ZY, Qu K and Liu C: Fork head box M1 regulates vascular endothelial growth factor-A expression to promote the angiogenesis and tumor cell growth of gallbladder cancer. World J Gastroenterol. 27:692–707. 2021. View Article : Google Scholar : PubMed/NCBI

87 

Zhang Y, Cheng C, Wang S, Xu M, Zhang D and Zeng W: Knockdown of FOXM1 inhibits activation of keloid fibroblasts and extracellular matrix production via inhibition of TGF-β1/Smad pathway. Life Sci. 232:1166372019. View Article : Google Scholar

88 

Mencalha AL, Binato R, Ferreira GM, Du Rocher B and Abdelhay E: Forkhead box M1 (FoxM1) gene is a new STAT3 transcriptional factor target and is essential for proliferation, survival and DNA repair of K562 cell line. PLoS One. 7:e481602012. View Article : Google Scholar : PubMed/NCBI

89 

Imai J, Katagiri H, Yamada T, Ishigaki Y, Suzuki T, Kudo H, Uno K, Hasegawa Y, Gao J, Kaneko K, et al: Regulation of pancreatic beta cell mass by neuronal signals from the liver. Science. 322:1250–1254. 2008. View Article : Google Scholar : PubMed/NCBI

90 

Frustaci A, Kajstura J, Chimenti C, Jakoniuk I, Leri A, Maseri A, Nadal-Ginard B and Anversa P: Myocardial cell death in human diabetes. Circ Res. 87:1123–1132. 2000. View Article : Google Scholar : PubMed/NCBI

91 

Dai Z, Zhu MM, Peng Y, Jin H, Machireddy N, Qian Z, Zhang X and Zhao YY: Endothelial and Smooth muscle cell interaction via FoxM1 Signaling mediates vascular remodeling and pulmonary hypertension. Am J Respir Crit Care Med. 198:788–802. 2018. View Article : Google Scholar : PubMed/NCBI

92 

Wang Y, Li Y, Feng J, Liu W, Li Y, Liu J, Yin Q, Lian H, Liu L and Nie Y: Mydgf promotes Cardiomyocyte proliferation and Neonatal Heart regeneration. Theranostics. 10:9100–9112. 2020. View Article : Google Scholar : PubMed/NCBI

93 

Eltoft A, Arntzen KA, Wilsgaard T, Mathiesen EB and Johnsen SH: Interleukin-6 is an independent predictor of progressive atherosclerosis in the carotid artery: The Tromso Study. Atherosclerosis. 271:1–8. 2018. View Article : Google Scholar : PubMed/NCBI

94 

Li H, Horke S and Forstermann U: Vascular oxidative stress, nitric oxide and atherosclerosis. Atherosclerosis. 237:208–219. 2014. View Article : Google Scholar : PubMed/NCBI

95 

Nahrendorf M and Swirski FK: Cholesterol, CCR2, and monocyte phenotypes in atherosclerosis. Eur Heart J. 38:1594–1596. 2017. View Article : Google Scholar : PubMed/NCBI

96 

Balli D, Ren X, Chou FS, Cross E, Zhang Y, Kalinichenko VV and Kalin TV: Foxm1 transcription factor is required for macrophage migration during lung inflammation and tumor formation. Oncogene. 31:3875–3888. 2012. View Article : Google Scholar :

97 

Lok GT, Chan DW, Liu VW, Hui WW, Leung TH, Yao KM and Ngan HY: Aberrant activation of ERK/FOXM1 signaling cascade triggers the cell migration/invasion in ovarian cancer cells. PLoS One. 6:e237902011. View Article : Google Scholar : PubMed/NCBI

98 

Xie Y, Cui D, Sui L, Xu Y, Zhang N, Ma Y, Li Y and Kong Y: Induction of forkhead box M1 (FoxM1) by EGF through ERK signaling pathway promotes trophoblast cell invasion. Cell Tissue Res. 362:421–430. 2015. View Article : Google Scholar : PubMed/NCBI

99 

Brem H and Tomic-Canic M: Cellular and molecular basis of wound healing in diabetes. J Clin Invest. 117:1219–1222. 2007. View Article : Google Scholar : PubMed/NCBI

100 

Armstrong DG, Swerdlow MA, Armstrong AA, Conte MS, Padula WV and Bus SA: Five year mortality and direct costs of care for people with diabetic foot complications are comparable to cancer. J Foot Ankle Res. 13:162020. View Article : Google Scholar : PubMed/NCBI

101 

Soo BP, Rajbhandari S, Egun A, Ranasinghe U, Lahart IM and Pappachan JM: Survival at 10 years following lower extremity amputations in patients with diabetic foot disease. Endocrine. 69:100–106. 2020. View Article : Google Scholar : PubMed/NCBI

102 

Smirnov A, Panatta E, Lena A, Castiglia D, Di Daniele N, Melino G and Candi E: FOXM1 regulates proliferation, senescence and oxidative stress in keratinocytes and cancer cells. Aging (Albany NY). 8:1384–1397. 2016. View Article : Google Scholar : PubMed/NCBI

103 

Brinkmann V, Reichard U, Goosmann C, Fauler B, Uhlemann Y, Weiss DS, Weinrauch Y and Zychlinsky A: Neutrophil extracellular traps kill bacteria. Science. 303:1532–1535. 2004. View Article : Google Scholar : PubMed/NCBI

104 

Lood C, Blanco LP, Purmalek MM, Carmona-Rivera C, De Ravin SS, Smith CK, Malech HL, Ledbetter JA, Elkon KB and Kaplan MJ: Neutrophil extracellular traps enriched in oxidized mitochondrial DNA are interferogenic and contribute to lupus-like disease. Nat Med. 22:146–153. 2016. View Article : Google Scholar : PubMed/NCBI

105 

Carrasco K, Boufenzer A, Jolly L, Le Cordier H, Wang G, Heck AJ, Cerwenka A, Vinolo E, Nazabal A, Kriznik A, et al: TREM-1 multimerization is essential for its activation on monocytes and neutrophils. Cell Mol Immunol. 16:460–472. 2019. View Article : Google Scholar :

106 

Palmer WC and Patel T: Are common factors involved in the pathogenesis of primary liver cancers? A meta-analysis of risk factors for intrahepatic cholangiocarcinoma. J Hepatol. 57:69–76. 2012. View Article : Google Scholar : PubMed/NCBI

107 

de Beer JC and Liebenberg L: Does cancer risk increase with HbA1c, independent of diabetes? Br J Cancer. 110:2361–2368. 2014. View Article : Google Scholar : PubMed/NCBI

108 

Hosokawa T, Kurosaki M, Tsuchiya K, Matsuda S, Muraoka M, Suzuki Y, Tamaki N, Yasui Y, Nakata T, Nishimura T, et al: Hyperglycemia is a significant prognostic factor of hepatocellular carcinoma after curative therapy. World J Gastroenterol. 19:249–257. 2013. View Article : Google Scholar : PubMed/NCBI

109 

Casagrande V, Mauriello A, Anemona L, Mavilio M, Iuliani G, De Angelis L, D'Onofrio M, Arisi I, Federici M and Menghini R: Timp3 deficiency affects the progression of DEN-related hepatocellular carcinoma during diet-induced obesity in mice. Acta Diabetol. 56:1265–1274. 2019. View Article : Google Scholar : PubMed/NCBI

110 

Andrei LG: Thiazole antibiotics siomycin a and thiostrepton inhibit the transcriptional activity of FOXM1. Front Oncol. 3:1502013.

111 

Shirakawa J, Fernandez M, Takatani T, El Ouaamari A, Jungtrakoon P, Okawa ER, Zhang W, Yi P, Doria A and Kulkarni RN: Insulin signaling regulates the FoxM1/PLK1/CENP-A pathway to promote adaptive pancreatic β cell proliferation. Cell Metab. 25:868–882 e5. 2017. View Article : Google Scholar

112 

Ma Y, Wang X, Peng Y and Ding X: Forkhead box O1 promotes INS-1 cell apoptosis by reducing the expression of CD24. Mol Med Rep. 13:2991–2998. 2016. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

November-2023
Volume 52 Issue 5

Print ISSN: 1107-3756
Online ISSN:1791-244X

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Zhao B, Li M, Su Y, Shan S, Qian W, Zhu D, Liu X and Zhang Z: Role of transcription factor FOXM1 in diabetes and its complications (Review). Int J Mol Med 52: 101, 2023
APA
Zhao, B., Li, M., Su, Y., Shan, S., Qian, W., Zhu, D. ... Zhang, Z. (2023). Role of transcription factor FOXM1 in diabetes and its complications (Review). International Journal of Molecular Medicine, 52, 101. https://doi.org/10.3892/ijmm.2023.5304
MLA
Zhao, B., Li, M., Su, Y., Shan, S., Qian, W., Zhu, D., Liu, X., Zhang, Z."Role of transcription factor FOXM1 in diabetes and its complications (Review)". International Journal of Molecular Medicine 52.5 (2023): 101.
Chicago
Zhao, B., Li, M., Su, Y., Shan, S., Qian, W., Zhu, D., Liu, X., Zhang, Z."Role of transcription factor FOXM1 in diabetes and its complications (Review)". International Journal of Molecular Medicine 52, no. 5 (2023): 101. https://doi.org/10.3892/ijmm.2023.5304