Open Access

Role of Takeda G protein‑coupled receptor 5 in microvascular endothelial cell dysfunction in diabetic retinopathy (Review)

  • Authors:
    • Miao Zhang
    • Zhenghao Dong
    • Wenkang Dong
    • Dongdong Zhou
    • Xiang Ren
  • View Affiliations

  • Published online on: September 15, 2022     https://doi.org/10.3892/etm.2022.11610
  • Article Number: 674
  • Copyright: © Zhang 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

Diabetic retinopathy (DR) is a frequent microvascular complication of advanced‑stage diabetes. Endothelial cell dysfunction (ED) induced by diabetes plays an important role in the development of DR. It is considered that inflammation and mitochondrial homeostasis are associated with the progression of ED. Takeda G protein‑coupled receptor 5 (TGR5) is a membrane receptor for bile acids (BAs) that plays an important role in regulating BA metabolism. Recent studies have shown that TGR5 is involved in regulating various mediators of ED and improving the dysfunction of vascular endothelial cells in DR; however, the exploration of specific related mechanisms remains an active research area in this field, which suggests that TGR5 may be one of the potential targets for the treatment of associated ED in DR. In the present review, the association between TGR5 and mitochondrial homeostasis was investigated. The extent of inflammation in DR‑induced ED was assessed to provide possible evidence for the development of targeted therapies against DR.

1. Introduction

Diabetic retinopathy (DR) is a common microvascular complication that presents at the late stages of diabetes. Approximately 80% of diabetic patients experience DR 20 years following onset, and its incidence is increasing worldwide (1,2). It has become one of the most important causes of blindness and visual impairment in working-age individuals (3,4). The initial stage of DR does not present with apparent symptoms; however, as the disease progresses, patients may experience blurred vision or even blindness (5,6). Early lesions in DR are characterized by loss of retinal capillary pericytes, resulting in increased vascular permeability, the presence of decellularized capillaries and microaneurysms, and rupture of the blood-retinal barrier (BRB) (7). Progression of DR to its later stage is followed by neocapillary proliferation, which significantly increases the likelihood of visual loss (8,9). Endothelial cell dysfunction (ED) is the key element to the development of microvascular lesions. Certain studies have shown that hyperglycemia-induced oxidative stress is increasing, which stimulates the inflammatory pathways and promotes vascular dysfunction of the retina leading to increased capillary permeability and vascular leakage (10). In addition, mitochondrial homeostasis is associated with ED (11). The enzyme, endothelial nitric oxide synthase (eNOS), also plays a vital role in maintaining the function of endothelial cells (ECs) (12).

Bile acids (BAs) are a class of endogenous molecules synthesized in the liver; they are present in the bile as ionic salts derived from the metabolism of cholesterol (13). The main role of cholesterol is to promote the digestion and absorption of lipids. In the case of diabetes mellitus (DM), lithocholic acids and deoxycholic acids, formed by the enterohepatic cycle of BAs, have a high affinity for Takeda G protein-coupled receptor 5 (TGR5); the BA-induced activation of TGR5 increases glucagon-like peptide-1 (GLP-1) and insulin release (14). In recent years, a high number of studies have shown that BAs can be used as a signaling molecule to bind to the corresponding receptors and participate in the regulation of various metabolic diseases (15,16). TGR5 is a widely studied signaling molecule involved in this process. TGR5 is expressed in a variety of tissues and organs, such as the liver, kidney, brain, and heart (17,18). It is also widely expressed in almost all types of ECs and is involved in the regulation of glucose and lipid metabolism processes present in various metabolic diseases, such as obesity, non-alcoholic fatty liver disease, and type 2 diabetes mellitus (T2DM) (19,20).

Currently, farnesoid X receptor (FXR) is mainly reported to be related to the function of macrovessels, while TGR5 is less relevant (21). In addition, as a small tissue in the eyeball, the blood vessels distributed on the retina are mainly microvessels. From an etiological point of view, the specific pathogenesis of DR remains unelucidated, but the current view accepted widely by researchers is that DR is a retinal microvascular complication induced by the long-term hyperglycemic environment. Therefore, in the present review, the role TGR5 plays in microvessels was investigated and an attempt was made to elucidate the underlying possible mechanisms.

To date, various studies on BAs and their receptors have implicated their possible roles in the regulation of EC function (22). Previously, it has been revealed that TGR5 is highly expressed in retinal microvascular ECs (23), which may produce BAs through the ‘alternative’ pathway (24). It has also been found that intermittent fasting increases the production of taurodeoxycholic acid (TUDCA), a metabolite of BAs in the retina, and protects retinal ECs to delay the progression of DR (23). A previous study has shown that TGR5 agonists are beneficial in diabetes and TGR5 has become a promising target for the treatment of this disease (25). Therefore, it was hypothesized that TGR5 activation may delay the progression of DR by improving ED, which plays a protective role in the retina; however, the underlying mechanism remains to be elucidated in further studies.

In the present review, the role of TGR5 in delaying the progression of DR was summarized by its effect on maintaining mitochondrial homeostasis and counteracting inflammation to protect ECs from damage. Therefore, the present study aimed to provide possible evidence for the application of the targeted therapy of DR.

2. DR and ED

ECs are a layer of squamous epithelial cells covering the inner surface of blood vessels, which constitute a barrier between blood vessels and tissues and control the transport of substances between tissues and blood vessels. ECs act as a metabolic interface between the blood and the tissues and are important in maintaining the stability of the intravascular environment (26). ED occurs when ECs are unable to maintain homeostasis of the vascular environment. It is a systemic pathological condition characterized by changes in the phenotype of ECs, which leads to diminished vasoconstriction and the formation of a proinflammatory and prothrombotic state (27). ED forms the basis of the chronic microvascular and macrovascular complications of diabetes. In recent years, significant progress has been made in understanding the mechanism of ED and its pathogenesis in patients with type 1 diabetes mellitus (T1DM) and T2DM. Several factors that cause ED have been identified and the common causes include hypoxia, aging, hyperglycemia, hypercholesterolemia, and hypertension (28). Previous studies have shown that pathophysiological processes caused by a high glucose environment found in diabetics, such as inflammation, oxidative stress, and endoplasmic reticulum (ER) stress are responsible for the continuous progression and aggravation of ED in the course of the disease (29). As a common microvascular complication in the late stage of diabetes, the risk factors for the development of DR are mainly related to the severity and exposure time of hyperglycemia, hypertension, and hyperlipidemia (30). ED is the pathological basis of diabetic microvascular complications and plays an important role in the pathological progression of DR. Progressive dysfunction of ECs will certainly lead to changes in morphological structures, such as capillary basement membrane thickening, perivascular cell loss, BRB damage, and neovascularization, which accelerates the progression of DR (31,32).

3. DR and mitochondrial homeostasis and inflammation

The specific mechanisms leading to DR have not been fully elucidated. However, disruption of mitochondrial homeostasis and inflammation are considered to be closely related to the pathogenesis of DR (33,34).

Diabetes can disturb mitochondrial dynamic homeostasis, causing impaired mitochondrial function, which in turn causes the development of related diseases (35,36). Under high glucose conditions, the electron flux through the electron transport chain increases, eventually leading to increased reactive oxygen species (ROS) production, which in turn causes retinal damage (37,38). The mitochondrial fusion division mechanisms are also compromised in diabetes; swollen retinal mitochondria decrease mitofusin 2 (Mfn2) expression and increase dynamin-related protein 1 (Drp1) expression (39). Decreased mitosis and inflammasome activation can be observed in the retina of diabetic patients (40), which further leads to deterioration of mitochondrial homeostasis.

In addition, several studies have implicated various systemic and local inflammatory factors in DR (41,42). Diabetes causes increased local and systemic expression of inflammatory cytokines, chemokines, and growth factors, all of which are involved in the development of DR (43-45). It has been shown that fortified extracts of red berries, ginkgo biloba leaves, and white willow bark containing carnosine and α-lipoic acid can significantly reduce cytokine levels in the retina and inhibit lipid peroxidation, which is associated with diabetes (46). Another study has demonstrated that curcumin can protect against high glucose-mediated retinal pigment epithelial cell injury due to induction of an anti-inflammatory pathway (47). Purinergic signaling has been shown to be a key factor in regulating the inflammatory status in different organ tissues. P2X purinergic receptor 7 (P2RX7) is a common purinergic ionotropic receptor; its activation leads to the release of proinflammatory mediators and the induction of cell damage. This receptor is considered to be a target for restoring BRB and reducing inflammation. It has been experimentally demonstrated that the inhibition or downregulation of P2X7R expression plays a protective role in inflammation-induced cell damage (48-50).

4. Structure of TGR5

TGR5, also known as G protein-coupled BA receptor 1 (GPBAR1) (51), belongs to the class A G-protein coupled receptor (GPCR) subfamily. The receptor comprises seven transmembrane helices (TMH1-7), three extracellular loops (ECL1-3), contributing to ligand binding, and three intracellular loops (ICL1-3) involved in mediating the signal to downstream signaling molecules (52). Substantial evidence has demonstrated that purinergic receptor P2Y9 in position 3.33 and chemokine receptor CXC3 in position 5.43 is involved in key ligand interactions. In addition, the receptor around Ser270 in TGR5 is able to recognize the acidic side chain of BAs, and the hydrophobic pocket hosting the C6 and C7 positions of the BA steroid nucleus was defined by hydrophobic residues including Phe96, Cys236, and Trp237(53). BA impacts TGR5 activity through those structures. TGR5 functions primarily through the TGR5-Gαs complex, a case in point is that the activation of TGR5 by oleanolic acid (OA) and INT-777 selectively activates Gαs and then the levels of intracellular TGR5-cyclin AMP (cAMP) will be increased (54). The interaction sites of TGR5 and Gαs are the intracellular terminal of TM6 on TGR5 and the GαsRas domain (mainly the C-terminal of the α5-helix). In addition, other than stabilizing the N-terminal α helix of Gαs, Gβ may also be involved in receptor binding (55) (Fig. 1).

5. TGR5 and ED

TGR5 is a common membrane receptor during BA metabolism and has been demonstrated to be expressed in a variety of tissues and organs (54). The role of TGR5 in regulating homeostatic metabolism is also well documented. A previous study has shown that TGR5 can delay the occurrence and development of portal hypertension by reversing ED (56). It was also found that activation of TGR5 could reverse the injury of liver sinusoidal ECs in a mouse model of cirrhosis and could reverse cardiovascular injury by reducing the secretion of inflammatory factors in aortic intimal cells (56,57). These studies indicate that activation of TGR5 may be a potential therapeutic strategy to delay ED caused by DR.

6. TGR5 and mitochondrial homeostasis

Previous studies have confirmed a close association between the damage of ECs and mitochondrial damage. Under damaged conditions, mitochondria generate large amounts of ROS, such as superoxide anion (O2-), hydrogen peroxide (H2O2), peroxyl radical (ROO·), and reactive hydroxyl radical (·OH), which are generally considered harmful to cells (58,59). Concomitantly, oxidative stress can cause changes in mitochondrial dynamics, such as activation of mitochondrial fission, inhibition of mitochondrial fusion, and an increase in the levels of mitophagy (60,61). Mitochondrial fusion involves three proteins, namely Mfn1, Mfn2, and optic atrophy 1 (OPA1), while mitochondrial fission is mediated by Drp1 and its receptors, including mitochondrial fission factor (MFF) and fission 1 (Fis1) (62,63). High levels of ROS can generate the release of inflammatory factors, such as vascular endothelial growth factor (VEGF), IL-1β, and TNF-α. For example, TNF-α can cause leakage of blood vessels by upregulating NK-κβ to activate ICAM-1(64) and induce EC damage. Numerous experiments have demonstrated that the production of a series of inflammatory cytokines and the changes in mitochondrial dynamics in diabetic rats contribute to retinal microvascular endothelial cell (RMEC) dysfunction in this animal model (65,66) (Fig. 2).

Mitochondria are the main site of energy production and play a crucial role in energy conversion and metabolism. In addition, mitochondria perform various functions that are essential for cell survival and have to maintain these processes and also adapt to the changing cellular environment. Mitochondria, as highly mobile double-membrane organelles, can form dynamic and extensive cellular networks that maintain homeostasis through fusion, fission, and mitophagy (62). Normally, fusion and division of mitochondria exist in a dynamic equilibrium. Mitochondrial dynamics are essential for the regulation of mitochondrial function and mitochondrial fragmentation has been shown to be involved in the induction of pathological processes including DM (67-69).

Substantial evidence has demonstrated that therapies that improve mitochondrial function can ameliorate damage to retinal ECs. D-Arg-dimethylTyr-Lys-Phe-NH2 (SS-31) is a mitochondria-targeted antioxidant peptide, which effectively reverses the decreased visual acuity in a streptozotocin-induced diabetic mouse model (70). Huang et al (71) demonstrated that diabetic rats treated with SS-31 exhibited improved retinal ganglion cell structure, thinner capillary basement membrane, and reduced inner BRB leakage. Therefore, the improvement of mitochondrial damage may become a new strategy to treat DR.

ECs rely on glycolysis for energy supply, which may be a misleading concept suggesting that adenosine triphosphate (ATP) derived from mitochondria has no important role in ECs (72). However, recent evidence suggests that while the energy requirements between ECs are not as large as those of cardiomyocytes and smooth muscle cells, intracellular ATP may play an important role in mediating the normal physiological functions of ECs (73). Mitochondrial oxidative phosphorylation plays an integral role in energy stores and mitochondrial dysfunction can enhance oxidative stress sensitivity and lead to EC death (65). Therefore, a decreased mitochondrial function also contributes to ED. Recently, it has been shown that EC injury can be delayed by reducing mitochondrial division and/or enhancing mitophagy via the activation of TGR5.

It has been shown that INT-777, an agonist of TGR5, prevents mitochondrial division by decreasing calcium concentration, attenuating protein kinase C (PKC) activation, and inhibiting the Ca2+-PKCδ/Drp1 pathway (65). It has also been found that PKCδ can lead to Drp1 phosphorylation and translocation of phosphorylated Drp1 to mitochondria can promote mitochondrial division (74). Intracellular calcium can activate calcineurin to promote mitochondrial division and induce activation of Ca2+/calmodulin-dependent protein kinase II, which can mediate p-S616 expression in Drp1 (75,76). Activation of TGR5 can reduce intracellular Ca2+ concentrations by blocking the influx of extracellular Ca2+, thereby inhibiting mitochondrial division. Concomitantly, decreased intracellular Ca2+ inhibits the extracellular regulated protein kinases (ERK1/2) signaling pathway, which can cause a decrease in Drp1 expression; this, in turn inhibits mitochondrial division and promotes Mfn1 oligomer formation, thereby promoting mitochondrial fusion (77-79).

Physiologically, a low number of damaged mitochondria are formed during mitochondrial fission, and damaged mitochondria are degraded by mitochondria-targeted autophagy termed mitophagy (80). TGR5 can activate the PTEN-induced kinase (PINK)/Parkin pathway and inhibit the PKCδ/Drp1-hexokinase (HK)2 pathway to enhance mitophagy. HK is a positive modulator of Parkin recruitment and glycolysis (81,82). As the major HK isoform in insulin-sensitive tissues including retinopathy, HK2 binds to voltage-dependent anion channels and localizes to the outer mitochondrial membrane. HK2 translocates from the mitochondria into the cytosol in response to high glucose conditions in diabetic mice. Treatment with INT-777 promotes recruitment of HK2 to the mitochondria and further activation of the PINK1/Parkin signaling pathway. The use of the Drp1 inhibitor Mdivi-1 can promote, in a similar manner, the translocation of HK2 from the cytosol to the mitochondria. This suggests a role for TGR5 in enhancing mitophagy and inhibiting cell division in vitro. In vivo, capillary degeneration and pericyte loss has been shown to be milder in TGR5-knockdown rats injected with the mitochondrial fission inhibitor Mdivi-1 or the mitophagy agonist rapamycin compared with that noted in control animals (65). In summary, TGR5 maintains mitochondrial homeostasis by reducing mitochondrial division and enhancing mitophagy, which in turn improves ED to delay the progression of DR (Fig. 3).

7. TGR5 and inflammation

DR is classified as a chronic low-level inflammatory process and accumulating evidence has shown that minor inflammation is responsible for the vasculopathy of DR. In the presence of oxidative stress caused by hyperglycemia, the levels of inflammatory factors, such as cytokines, VEGF, IL-1β, and TNF-α in the serum and local microenvironment are increased. This increase in inflammatory factors leads to retinopathy (83). Previous studies have demonstrated that activation of the TGR5 receptor can delay the production of IL-1, TNF-α, and other inflammatory factors by macrophages; it may also reduce the production of proinflammatory factors by inhibiting the Toll-like receptor (TLR)4/NK-κβ pathway and can play a role in inhibiting the phosphorylation of STAT3 (21,84,85). Among all inflammatory factors, TNF-α was first implicated in the progression of insulin resistance, as well as in abnormal glucose metabolism associated with T2DM. Therefore, the association between inflammation and DR was assessed with regard to the contribution of TNF-α.

It has been reported that TNF-α can activate various caspases leading to apoptosis of inflammatory cells in the chronic inflammatory response. TNF-α can also activate NK-κβ, thereby causing an upregulation in the expression levels of related genes involved in inflammation and resulting in increased intercellular adhesion molecule 1 (ICAM-1) synthesis. A large amount of ICAM-1 will damage vascular ECs following binding to activated leukocytes, resulting in vascular leakage at the corresponding site (64). Several lines of evidence suggest that the elevation of TNF-α is significantly associated with diabetic angiopathy in diabetic complications. The experiments indicated higher TNF-α levels in the serum of diabetic rats than those noted in normal mice in vivo. Following treatment with apigenin and ramipril, the increase in the levels of TNF-α was inhibited. Moreover, glomerular hypertrophy, fibrosis, and matrix expansion were improved, and the degree of inflammation was reduced in diabetic rats (86). In vitro, it has been demonstrated that the mRNA expression and secretion of TNF-α are markedly upregulated in human glomerular EC cells (HRGECs) treated with high concentrations of glucose (87). A recent study has also demonstrated that TNF-α levels are elevated in the early stages of retinopathy and remain high throughout the process; therefore, it is speculated that TNF-α can be used as a marker to predict DR (88).

TGR5 has been found to regulate TNF-α by modulating the Rho/Rho-associated protein kinase (ROCK) signaling pathway. As an agonist of TGR5, INT-777 can block TNF-α-induced RMEC proliferation and migration and inhibit the effect of TNF-α on promoting vascular permeability (66,89). In addition, previous evidence has shown that TGR5 agonists can upregulate IL-10 expression to exert anti-inflammatory and immunosuppressive effects and reduce the expression of the proinflammatory cytokines IL-1β, IL-6, and TNF-α by activating the TGR5-cAMP-protein kinase A (PKA) signaling pathway (90,91). In addition to the two classical signaling pathways described above, GLP-1, an intestinal hormone with a short half-life, has been shown to inhibit inflammation and improve EC function (92). A study has shown that GLP-1 can delay the damage of ECs by inhibiting NF-κB, which in turn inhibits the secretion of inflammatory factors, such as IL-1β, IL-6, and TNF-α (93). While various studies confirm that GLP-1 is one of the targets of TGR5; its secretion is dependent on TGR5 (93,94). Therefore, it was speculated that activation of TGR5 may be an effective way to stimulate GLP-1 secretion and delay RMEC injury (Fig. 4).

8. Discussion and future perspectives

As previously mentioned, oxidative stress, inflammation, and mitochondrial damage are important mechanisms of DR that lead to EC damage. The activation of TGR5 can reduce TNF-α expression via the Rho/ROCK pathway as well as promote the secretion of GLP-1 (66,94). TGR5 affects microsomal kinetics by inhibiting the Ca2+-PKCδ/Drp1 pathway, upregulating the PINK/Parkin pathway, and regulating the PKCδ/Drp1-HK2 pathway to alter DR. Therefore, the ability of TGR5 to ameliorate EC function may be one of the potential mechanisms responsible for its inhibitory effect on DR.

In addition to the aforementioned mechanisms, activation of TGR5 stimulates vascular ECs to produce eNOS to maintain vascular health and function; this mechanism has also the potential to improve DR (95). Previous studies have shown that activation of TGR5 can effectively increase eNOS expression; and its expression level is increased through the bile salt-TGR5-cAMP pathway and the TGR5-GLP-1-PI3K-eNOS pathway. (17,27). In addition, a study has also shown that taurolithocholic acids (TLCAs), taurocholic acid (TCA), and taurochenodeoxycholic acid (TCDCA) as agonists of eNOS has been shown to elevate eNOS expression and Ser1177 phosphorylation of this enzyme, leading to increased nitric oxide (NO) production (96). The increase in NO production can effectively protect the vascular endothelium.

Exchange proteins directly activated by cAMP (EPACs), consisting of Epac1 and Epac2, are cAMP mediators independent of PKA. As a mediator of cAMP, Epacs take part in numerous biological functions (97,98). Increasing studies in recent years have demonstrated the role cAMP/Epac signaling plays in endothelial cell barrier function (99,100). It has been found that Epac-1 expression is significantly reduced in mouse models of DR, suggesting that Epac-1 is a critical regulator of endothelial function in diabetic microangiopathy involving endothelial dysfunction associated with hypoxia. Activation of Epac-1 by forskolin or the cAMP analog 8-pCPT reduces its sensitivity to oxidative stress, restores the endothelial permeability barrier, rescues NO production by eNOS and inhibits ROS formation (101), suggesting that Epac-1 may be a potential target for the treatment of ED during DR. It has also been revealed that activation of Epac inhibits VEGF receptor signaling through the Ras/MEK/ERK pathway to improve BRB permeability (102). In addition, Epac has also been demonstrated to reduce inflammatory mediators in retinal endothelial cells, potentially mediating anti-inflammatory responses in endothelial cells (103). In the present review, it was indicated that TGR5 can activate cAMP, from which it can be speculated that TGR5, after activating cAMP, may improve retinal endothelial cell function through cAMP/Epac signaling, thereby delaying the progression of DR.

To date, insufficient evidence has been reported supporting the notion that TGR5 can delay vascular endothelial injury in DR through ER stress. Therefore, it is possible that a new mechanism may be responsible for this process. Achieving a relatively balanced state of mitochondria by maintaining the homeostasis of the ER has the potential to be a novel mechanism by which TGR5 delays EC injury. ER stress, such as interference in Ca2+ homeostasis, redox imbalance, and defects in protein folding can cause disorders in ECs (104). In ER stress, the unfolded protein response (UPR) can be triggered and the UPR is an adaptive process to restore ER stress. It has been shown that oral administration of TUDCA can reduce ED triggered by hyperglycemia. Moreover, activation of the UPR by the use of the chemical chaperone TUDCA can alleviate the glucose-induced increase in inflammatory cytokines and endothelin-1, as well as the decrease in NO levels (105,106). It has been demonstrated that ER stress is associated with TGR5 and that TGR5 mRNA levels are upregulated in skeletal myotubes in response to the UPR inducers thapsigargin (ER-specific Ca-ATPase inhibitor) and tunicamycin (N-glycosylation inhibitor), demonstrating that TGR5 is a novel UPR target gene (107). Additional studies have demonstrated that TUDCA can reduce ER stress by stimulating the TGR5 signaling pathway (108,109). It has been shown that upregulation of TGR5 expression inhibits the TLR4/NF-κB pathway to reduce oxidative stress, which may delay endothelial injury (85). TGR5 may alleviate ER stress through the protein kinase R (PKR)-like endoplasmic reticulum kinase (PERK)/eukaryotic initiation factor 2(eIF2)/NF-κB pathway and activating transcription factor 4-CCAAT-enhancer-binding protein homologous protein (ATF4-CHOP); therefore, these targets have the potential to be used for delaying necrosis of diabetic RMECs (104).

Since the activation of the TGR5 receptor improves EC injury caused by mitochondrial injury, oxidative stress, inflammatory factors, and ER stress, it may play a role in delaying DR-induced ED. Therefore, the TGR5 receptor can be used as a new target to ameliorate DR. Quinoa can cause an upregulation of the expression of GLP-1 via increased expression levels of TGR5 and it may be useful in the treatment of DR (85). Therefore, it is possible that certain components in Chenopodium album may act as agonists of TGR5. This application can be supported further by in vivo studies and clinical trials in humans.

Acknowledgements

Not applicable.

Funding

Funding: The present review was supported by the Natural Science Foundation of Liaoning Province (grant no. 2020-BS-189), the Basic Scientific Research Projects of Liaoning Provincial Education Department (grant no. LQ2017005) and the Liaoning Provincial Program for Top Discipline of Basic Medical Sciences (grant nos. 2020-BS-189 and LQ2017005).

Availability of data and materials

Not applicable.

Authors' contributions

MZ conceived and designed the article. MZ and ZD prepared and wrote the manuscript. MZ, ZD and WD performed a literature search, selected the studies to be included and drew the figures. MZ, ZD, DZ and XR revised the manuscript. DZ retrieved and analyzed relevant documents. 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.

References

1 

Williams R, Airey M, Baxter H, Forrester J, Kennedy-Martin T and Girach A: Epidemiology of diabetic retinopathy and macular oedema: A systematic review. Eye (Lond). 18:963–983. 2004.PubMed/NCBI View Article : Google Scholar

2 

Simó R and Hernández C: Prevention and treatment of diabetic retinopathy: Evidence from large, randomized trials. The emerging role of fenofibrate. Rev Recent Clin Trials. 7:71–80. 2012.PubMed/NCBI View Article : Google Scholar

3 

Singh R, Barden A, Mori T and Beilin L: Advanced glycation end-products: A review. Diabetologia. 44:129–146. 2001.PubMed/NCBI View Article : Google Scholar

4 

Pardue MT and Allen RS: Neuroprotective strategies for retinal disease. Prog Retin Eye Res. 65:50–76. 2018.PubMed/NCBI View Article : Google Scholar

5 

Kollias AN and Ulbig MW: Diabetic retinopathy: Early diagnosis and effective treatment. Dtsch Arztebl Int. 107:75–83; quiz 84. 2010.PubMed/NCBI View Article : Google Scholar

6 

Moreno A, Lozano M and Salinas P: Diabetic retinopathy. Nutr Hosp. 28 (Suppl 2):S53–S56. 2013.PubMed/NCBI View Article : Google Scholar

7 

Wong TY, Cheung CM, Larsen M, Sharma S and Simó R: Diabetic retinopathy. Nat Rev Dis Primers. 2(16012)2016.PubMed/NCBI View Article : Google Scholar

8 

Henriques J, Vaz-Pereira S, Nascimento J and Rosa PC: Diabetic eye disease. Acta Med Port. 28:107–113. 2015.PubMed/NCBI(In Portuguese).

9 

Lechner J, O'Leary OE and Stitt AW: The pathology associated with diabetic retinopathy. Vision Res. 139:7–14. 2017.PubMed/NCBI View Article : Google Scholar

10 

Shafabakhsh R, Aghadavod E, Mobini M, Heidari-Soureshjani R and Asemi Z: Association between microRNAs expression and signaling pathways of inflammatory markers in diabetic retinopathy. J Cell Physiol. 234:7781–7787. 2019.PubMed/NCBI View Article : Google Scholar

11 

Singh LP, Yumnamcha T and Devi TS: Mitophagy, ferritinophagy and ferroptosis in retinal pigment epithelial cells under high glucose conditions: Implications for Diabetic retinopathy and age-related retinal diseases. JOJ Ophthalmol. 8:77–85. 2021.PubMed/NCBI

12 

Meza CA, La Favor JD, Kim DH and Hickner RC: Endothelial dysfunction: Is There a Hyperglycemia-induced imbalance of NOX and NOS? Int J Mol Sci. 20(3775)2019.PubMed/NCBI View Article : Google Scholar

13 

Maldonado-Valderrama J, Wilde P, Macierzanka A and Mackie A: The role of bile salts in digestion. Adv Colloid Interface Sci. 165:36–46. 2011.PubMed/NCBI View Article : Google Scholar

14 

van Nierop FS, Scheltema MJ, Eggink HM, Pols TW, Sonne DP, Knop FK and Soeters MR: Clinical relevance of the bile acid receptor TGR5 in metabolism. Lancet Diabetes Endocrinol. 5:224–233. 2017.PubMed/NCBI View Article : Google Scholar

15 

de Boer JF, Bloks VW, Verkade E, Heiner-Fokkema MR and Kuipers F: New insights in the multiple roles of bile acids and their signaling pathways in metabolic control. Curr Opin Lipidol. 29:194–202. 2018.PubMed/NCBI View Article : Google Scholar

16 

Molinaro A, Wahlström A and Marschall HU: Role of bile acids in metabolic control. Trends Endocrinol Metab. 29:31–41. 2018.PubMed/NCBI View Article : Google Scholar

17 

Thomas C, Gioiello A, Noriega L, Strehle A, Oury J, Rizzo G, Macchiarulo A, Yamamoto H, Mataki C, Pruzanski M, et al: TGR5-mediated bile acid sensing controls glucose homeostasis. Cell Metab. 10:167–177. 2009.PubMed/NCBI View Article : Google Scholar

18 

Li S, Qiu M, Kong Y, Zhao X, Choi HJ, Reich M, Bunkelman BH, Liu Q, Hu S, Han M, et al: Bile Acid G protein-coupled membrane receptor TGR5 Modulates Aquaporin 2-Mediated water homeostasis. J Am Soc Nephrol. 29:2658–2670. 2018.PubMed/NCBI View Article : Google Scholar

19 

Li T and Chiang JY: Bile acid signaling in metabolic disease and drug therapy. Pharmacol Rev. 66:948–983. 2014.PubMed/NCBI View Article : Google Scholar

20 

Chávez-Talavera O, Tailleux A, Lefebvre P and Staels B: Bile acid control of metabolism and inflammation in obesity, type 2 diabetes, dyslipidemia, and nonalcoholic fatty liver disease. Gastroenterology. 152:1679–1694.e3. 2017.PubMed/NCBI View Article : Google Scholar

21 

Voiosu A, Wiese S, Voiosu T, Bendtsen F and Møller S: Bile acids and cardiovascular function in cirrhosis. Liver Int. 37:1420–1430. 2017.PubMed/NCBI View Article : Google Scholar

22 

Wang XX, Wang D, Luo Y, Myakala K, Dobrinskikh E, Rosenberg AZ, Levi J, Kopp JB, Field A, Hill A, et al: FXR/TGR5 dual agonist prevents progression of nephropathy in diabetes and obesity. J Am Soc Nephrol. 29:118–137. 2018.PubMed/NCBI View Article : Google Scholar

23 

Beli E, Yan Y, Moldovan L, Vieira CP, Gao R, Duan Y, Prasad R, Bhatwadekar A, White FA, Townsend SD, et al: Restructuring of the gut microbiome by intermittent fasting prevents retinopathy and prolongs survival in db/db Mice. Diabetes. 67:1867–1879. 2018.PubMed/NCBI View Article : Google Scholar

24 

Ren S, Hylemon P, Marques D, Hall E, Redford K, Gil G and Pandak WM: Effect of increasing the expression of cholesterol transporters (StAR, MLN64, and SCP-2) on bile acid synthesis. J Lipid Res. 45:2123–2131. 2004.PubMed/NCBI View Article : Google Scholar

25 

Pellicciari R, Gioiello A, Macchiarulo A, Thomas C, Rosatelli E, Natalini B, Sardella R, Pruzanski M, Roda A, Pastorini E, et al: Discovery of 6alpha-ethyl-23(S)-methylcholic acid (S-EMCA, INT-777) as a potent and selective agonist for the TGR5 receptor, a novel target for diabesity. J Med Chem. 52:7958–7961. 2009.PubMed/NCBI View Article : Google Scholar

26 

Galley HF and Webster NR: Physiology of the endothelium. Br J Anaesth. 93:105–113. 2004.PubMed/NCBI View Article : Google Scholar

27 

Cai Z, Yuan S, Zhong Y, Deng L, Li J, Tan X and Feng J: Amelioration of Endothelial dysfunction in diabetes: Role of takeda G protein-coupled receptor 5. Front Pharmacol. 12(637051)2021.PubMed/NCBI View Article : Google Scholar

28 

Poredos P, Poredos AV and Gregoric I: Endothelial dysfunction and its clinical implications. Angiology. 72:604–615. 2021.PubMed/NCBI View Article : Google Scholar

29 

Basha B, Samuel SM, Triggle CR and Ding H: Endothelial dysfunction in diabetes mellitus: Possible involvement of endoplasmic reticulum stress? Exp Diabetes Res. 2012(481840)2012.PubMed/NCBI View Article : Google Scholar

30 

Sorrentino FS, Matteini S, Bonifazzi C, Sebastiani A and Parmeggiani F: Diabetic retinopathy and endothelin system: Microangiopathy versus endothelial dysfunction. Eye (Lond). 32:1157–1163. 2018.PubMed/NCBI View Article : Google Scholar

31 

Brownlee M: The pathobiology of diabetic complications: A unifying mechanism. Diabetes. 54:1615–1625. 2005.PubMed/NCBI View Article : Google Scholar

32 

Fu D, Yu JY, Yang S, Wu M, Hammad SM, Connell AR, Du M, Chen J and Lyons TJ: Survival or death: A dual role for autophagy in stress-induced pericyte loss in diabetic retinopathy. Diabetologia. 59:2251–2261. 2016.PubMed/NCBI View Article : Google Scholar

33 

Kowluru RA: Mitochondrial stability in diabetic retinopathy: Lessons learned from epigenetics. Diabetes. 68:241–247. 2019.PubMed/NCBI View Article : Google Scholar

34 

Forrester JV, Kuffova L and Delibegovic M: The role of inflammation in diabetic retinopathy. Front Immunol. 11(583687)2020.PubMed/NCBI View Article : Google Scholar

35 

Liang Q and Kobayashi S: Mitochondrial quality control in the diabetic heart. J Mol Cell Cardiol. 95:57–69. 2016.PubMed/NCBI View Article : Google Scholar

36 

Williams M and Caino MC: Mitochondrial dynamics in type 2 diabetes and cancer. Front Endocrinol (Lausanne). 9(211)2018.PubMed/NCBI View Article : Google Scholar

37 

Madsen-Bouterse SA, Mohammad G, Kanwar M and Kowluru RA: Role of mitochondrial DNA damage in the development of diabetic retinopathy, and the metabolic memory phenomenon associated with its progression. Antioxid Redox Signal. 13:797–805. 2010.PubMed/NCBI View Article : Google Scholar

38 

Kowluru RA and Mishra M: Regulation of matrix metalloproteinase in the pathogenesis of diabetic retinopathy. Prog Mol Biol Transl Sci. 148:67–85. 2017.PubMed/NCBI View Article : Google Scholar

39 

Zhong Q and Kowluru RA: Diabetic retinopathy and damage to mitochondrial structure and transport machinery. Invest Ophthalmol Vis Sci. 52:8739–8746. 2011.PubMed/NCBI View Article : Google Scholar

40 

Singh LP, Devi TS and Yumnamcha T: The role of txnip in mitophagy dysregulation and inflammasome activation in diabetic retinopathy: A new perspective. JOJ Ophthalmol. 4(10)2017.PubMed/NCBI View Article : Google Scholar

41 

Noda K, Nakao S, Ishida S and Ishibashi T: Leukocyte adhesion molecules in diabetic retinopathy. J Ophthalmol. 2012(279037)2012.PubMed/NCBI View Article : Google Scholar

42 

Kaštelan S, Tomić M, Gverović Antunica A, Salopek Rabatić J and Ljubić S: Inflammation and pharmacological treatment in diabetic retinopathy. Mediators Inflamm. 2013(213130)2013.PubMed/NCBI View Article : Google Scholar

43 

Tang J and Kern TS: Inflammation in diabetic retinopathy. Prog Retin Eye Res. 30:343–358. 2011.PubMed/NCBI View Article : Google Scholar

44 

Shabab T, Khanabdali R, Moghadamtousi SZ, Kadir HA and Mohan G: Neuroinflammation pathways: A general review. Int J Neurosci. 127:624–633. 2017.PubMed/NCBI View Article : Google Scholar

45 

Rübsam A, Parikh S and Fort PE: Role of inflammation in diabetic retinopathy. Int J Mol Sci. 19(942)2018.PubMed/NCBI View Article : Google Scholar

46 

Bucolo C, Marrazzo G, Platania CB, Drago F, Leggio GM and Salomone S: Fortified extract of red berry, Ginkgo biloba, and white willow bark in experimental early diabetic retinopathy. J Diabetes Res. 2013(432695)2013.PubMed/NCBI View Article : Google Scholar

47 

Bucolo C, Drago F, Maisto R, Romano GL, D'Agata V, Maugeri G and Giunta S: Curcumin prevents high glucose damage in retinal pigment epithelial cells through ERK1/2-mediated activation of the Nrf2/HO-1 pathway. J Cell Physiol. 234:17295–17304. 2019.PubMed/NCBI View Article : Google Scholar

48 

Platania CBM, Lazzara F, Fidilio A, Fresta CG, Conti F, Giurdanella G, Leggio GM, Salomone S, Drago F and Bucolo C: Blood-retinal barrier protection against high glucose damage: The role of P2X7 receptor. Biochem Pharmacol. 168:249–258. 2019.PubMed/NCBI View Article : Google Scholar

49 

Tassetto M, Scialdone A, Solini A and Di Virgilio F: The P2X7 receptor: A promising pharmacological target in diabetic retinopathy. Int J Mol Sci. 22(7110)2021.PubMed/NCBI View Article : Google Scholar

50 

Platania CBM, Drago F and Bucolo C: The P2X7 receptor as a new pharmacological target for retinal diseases. Biochem Pharmacol. 198(114942)2022.PubMed/NCBI View Article : Google Scholar

51 

Maruyama T, Miyamoto Y, Nakamura T, Tamai Y, Okada H, Sugiyama E, Nakamura T, Itadani H and Tanaka K: Identification of membrane-type receptor for bile acids (M-BAR). Biochem Biophys Res Commun. 298:714–719. 2002.PubMed/NCBI View Article : Google Scholar

52 

Hov JR, Keitel V, Laerdahl JK, Spomer L, Ellinghaus E, ElSharawy A, Melum E, Boberg KM, Manke T, Balschun T, et al: Mutational characterization of the bile acid receptor TGR5 in primary sclerosing cholangitis. PLoS One. 5(e12403)2010.PubMed/NCBI View Article : Google Scholar

53 

Macchiarulo A, Gioiello A, Thomas C, Pols TW, Nuti R, Ferrari C, Giacchè N, De Franco F, Pruzanski M, Auwerx J, et al: Probing the binding site of bile acids in TGR5. ACS Med Chem Lett. 4:1158–1162. 2013.PubMed/NCBI View Article : Google Scholar

54 

Guo C, Chen WD and Wang YD: TGR5, not only a metabolic regulator. Front Physiol. 7(646)2016.PubMed/NCBI View Article : Google Scholar

55 

Chen G, Wang X, Ge Y, Ma L, Chen Q, Liu H, Du Y, Ye RD, Hu H and Ren R: Cryo-EM structure of activated bile acids receptor TGR5 in complex with stimulatory G protein. Signal Transduct Target Ther. 5(142)2020.PubMed/NCBI View Article : Google Scholar

56 

Renga B, Cipriani S, Carino A, Simonetti M, Zampella A and Fiorucci S: Reversal of Endothelial Dysfunction by GPBAR1 agonism in portal hypertension involves a AKT/FOXOA1 dependent regulation of H2S generation and endothelin-1. PLoS One. 10(e0141082)2015.PubMed/NCBI View Article : Google Scholar

57 

Carino A, Marchianò S, Biagioli M, Bucci M, Vellecco V, Brancaleone V, Fiorucci C, Zampella A, Monti MC, Distrutti E and Fiorucci S: Agonism for the bile acid receptor GPBAR1 reverses liver and vascular damage in a mouse model of steatohepatitis. FASEB J. 33:2809–2822. 2019.PubMed/NCBI View Article : Google Scholar

58 

Poprac P, Jomova K, Simunkova M, Kollar V, Rhodes CJ and Valko M: Targeting free radicals in oxidative stress-related human diseases. Trends Pharmacol Sci. 38:592–607. 2017.PubMed/NCBI View Article : Google Scholar

59 

Prasad S, Gupta SC and Tyagi AK: Reactive oxygen species (ROS) and cancer: Role of antioxidative nutraceuticals. Cancer Lett. 387:95–105. 2017.PubMed/NCBI View Article : Google Scholar

60 

Shutt T, Geoffrion M, Milne R and McBride HM: The intracellular redox state is a core determinant of mitochondrial fusion. EMBO Rep. 13:909–915. 2012.PubMed/NCBI View Article : Google Scholar

61 

Sabouny R, Fraunberger E, Geoffrion M, Ng AC, Baird SD, Screaton RA, Milne R, McBride HM and Shutt TE: The Keap1-Nrf2 stress response pathway promotes mitochondrial hyperfusion through degradation of the mitochondrial fission protein Drp1. Antioxid Redox Signal. 27:1447–1459. 2017.PubMed/NCBI View Article : Google Scholar

62 

Ferrington DA, Fisher CR and Kowluru RA: Mitochondrial defects drive degenerative retinal diseases. Trends Mol Med. 26:105–118. 2020.PubMed/NCBI View Article : Google Scholar

63 

Sabouny R and Shutt TE: Reciprocal regulation of mitochondrial fission and fusion. Trends Biochem Sci. 45:564–577. 2020.PubMed/NCBI View Article : Google Scholar

64 

Lutty GA: Effects of diabetes on the eye. Invest Ophthalmol Vis Sci. 54:ORSF81–ORSF87. 2013.PubMed/NCBI View Article : Google Scholar

65 

Zhang MY, Zhu L, Zheng X, Xie TH, Wang W, Zou J, Li Y, Li HY, Cai J, Gu S, et al: TGR5 activation ameliorates mitochondrial homeostasis via regulating the PKCδ/Drp1-HK2 signaling in diabetic retinopathy. Front Cell Dev Biol. 9(759421)2022.PubMed/NCBI View Article : Google Scholar

66 

Zhu L, Wang W, Xie TH, Zou J, Nie X, Wang X, Zhang MY, Wang ZY, Gu S, Zhuang M, et al: TGR5 receptor activation attenuates diabetic retinopathy through suppression of RhoA/ROCK signaling. FASEB J. 34:4189–4203. 2020.PubMed/NCBI View Article : Google Scholar

67 

Mishra P and Chan DC: Metabolic regulation of mitochondrial dynamics. J Cell Biol. 212:379–387. 2016.PubMed/NCBI View Article : Google Scholar

68 

Rovira-Llopis S, Bañuls C, Diaz-Morales N, Hernandez-Mijares A, Rocha M and Victor VM: Mitochondrial dynamics in type 2 diabetes: Pathophysiological implications. Redox Biol. 11:637–645. 2017.PubMed/NCBI View Article : Google Scholar

69 

Huang M, Wei R, Wang Y, Su T, Li P and Chen X: The uremic toxin hippurate promotes endothelial dysfunction via the activation of Drp1-mediated mitochondrial fission. Redox Biol. 16:303–313. 2018.PubMed/NCBI View Article : Google Scholar

70 

Alam NM, Mills WC IV, Wong AA, Douglas RM, Szeto HH and Prusky GT: A mitochondrial therapeutic reverses visual decline in mouse models of diabetes. Dis Model Mech. 8:701–710. 2015.PubMed/NCBI View Article : Google Scholar

71 

Huang J, Li X, Li M, Li J, Xiao W, Ma W, Chen X, Liang X, Tang S and Luo Y: Mitochondria-targeted antioxidant peptide SS31 protects the retinas of diabetic rats. Curr Mol Med. 13:935–945. 2013.PubMed/NCBI View Article : Google Scholar

72 

Dikalov SI, Nazarewicz RR, Bikineyeva A, Hilenski L, Lassègue B, Griendling KK, Harrison DG and Dikalova AE: Nox2-induced production of mitochondrial superoxide in angiotensin II-mediated endothelial oxidative stress and hypertension. Antioxid Redox Signal. 20:281–294. 2014.PubMed/NCBI View Article : Google Scholar

73 

Verónica Donoso M, Hernández F, Villalón T, Acuña-Castillo C and Pablo Huidobro-Toro J: Pharmacological dissection of the cellular mechanisms associated to the spontaneous and the mechanically stimulated ATP release by mesentery endothelial cells: Roles of thrombin and TRPV. Purinergic Signal. 14:121–139. 2018.PubMed/NCBI View Article : Google Scholar

74 

Chen C, Huang J, Shen J and Bai Q: Quercetin improves endothelial insulin sensitivity in obese mice by inhibiting Drp1 phosphorylation at serine 616 and mitochondrial fragmentation. Acta Biochim Biophys Sin (Shanghai). 51:1250–1257. 2019.PubMed/NCBI View Article : Google Scholar

75 

Slupe AM, Merrill RA, Flippo KH, Lobas MA, Houtman JC and Strack S: A calcineurin docking motif (LXVP) in dynamin-related protein 1 contributes to mitochondrial fragmentation and ischemic neuronal injury. J Biol Chem. 288:12353–12365. 2013.PubMed/NCBI View Article : Google Scholar

76 

Bo T, Yamamori T, Suzuki M, Sakai Y, Yamamoto K and Inanami O: Calmodulin-dependent protein kinase II (CaMKII) mediates radiation-induced mitochondrial fission by regulating the phosphorylation of dynamin-related protein 1 (Drp1) at serine 616. Biochem Biophys Res Commun. 495:1601–1607. 2018.PubMed/NCBI View Article : Google Scholar

77 

Cho B, Cho HM, Jo Y, Kim HD, Song M, Moon C, Kim H, Kim K, Sesaki H, Rhyu IJ, et al: Constriction of the mitochondrial inner compartment is a priming event for mitochondrial division. Nat Commun. 8(15754)2017.PubMed/NCBI View Article : Google Scholar

78 

Cook SJ, Stuart K, Gilley R and Sale MJ: Control of cell death and mitochondrial fission by ERK1/2 MAP kinase signalling. FEBS J. 284:4177–4195. 2017.PubMed/NCBI View Article : Google Scholar

79 

Chakrabarti R, Ji WK, Stan RV, de Juan Sanz J, Ryan TA and Higgs HN: INF2-mediated actin polymerization at the ER stimulates mitochondrial calcium uptake, inner membrane constriction, and division. J Cell Biol. 217:251–268. 2018.PubMed/NCBI View Article : Google Scholar

80 

Schmukler E, Solomon S, Simonovitch S, Goldshmit Y, Wolfson E, Michaelson DM and Pinkas-Kramarski R: Altered mitochondrial dynamics and function in APOE4-expressing astrocytes. Cell Death Dis. 11(578)2020.PubMed/NCBI View Article : Google Scholar

81 

Xu S and Herschman HR: A Tumor agnostic therapeutic strategy for hexokinase 1-Null/Hexokinase 2-positive cancers. Cancer Res. 79:5907–5914. 2019.PubMed/NCBI View Article : Google Scholar

82 

Li M, Shao J, Guo Z, Jin C, Wang L, Wang F, Jia Y, Zhu Z, Zhang Z, Zhang F, et al: Novel mitochondrion-targeting copper(II) complex induces HK2 malfunction and inhibits glycolysis via Drp1-mediating mitophagy in HCC. J Cell Mol Med. 24:3091–3107. 2020.PubMed/NCBI View Article : Google Scholar

83 

Clausell N, Kalil P, Biolo A, Molossi S and Azevedo M: Increased expression of tumor necrosis factor-alpha in diabetic macrovasculopathy. Cardiovasc Pathol. 8:145–151. 1999.PubMed/NCBI View Article : Google Scholar

84 

Zhou X and Guan Z, Jin X, Zhao J, Chen G, Ding J, Ren Y, Zhai X, Zhou Q and Guan Z: Reversal of alopecia areata, osteoporosis follow treatment with activation of Tgr5 in mice. Biosci Rep. 41(BSR20210609)2021.PubMed/NCBI View Article : Google Scholar

85 

Wang TY, Tao SY, Wu YX, An T, Lv BH, Liu JX, Liu YT and Jiang GJ: Quinoa Reduces High-Fat diet-induced obesity in mice via potential microbiota-gut-brain-liver interaction mechanisms. Microbiol Spectr. 10(e0032922)2022.PubMed/NCBI View Article : Google Scholar

86 

Malik S, Suchal K, Khan SI, Bhatia J, Kishore K, Dinda AK and Arya DS: Apigenin ameliorates streptozotocin-induced diabetic nephropathy in rats via MAPK-NF-κB-TNF-α and TGF-β1-MAPK-fibronectin pathways. Am J Physiol Renal Physiol. 313:F414–F422. 2017.PubMed/NCBI View Article : Google Scholar

87 

Xiang E, Han B, Zhang Q, Rao W, Wang Z, Chang C, Zhang Y, Tu C, Li C and Wu D: Human umbilical cord-derived mesenchymal stem cells prevent the progression of early diabetic nephropathy through inhibiting inflammation and fibrosis. Stem Cell Res Ther. 11(336)2020.PubMed/NCBI View Article : Google Scholar

88 

Khaloo P, Qahremani R, Rabizadeh S, Omidi M, Rajab A, Heidari F, Farahmand G, Bitaraf M, Mirmiranpour H, Esteghamati A and Nakhjavani M: Nitric oxide and TNF-α are correlates of diabetic retinopathy independent of hs-CRP and HbA1c. Endocrine. 69:536–541. 2020.PubMed/NCBI View Article : Google Scholar

89 

Mikelis CM, Simaan M, Ando K, Fukuhara S, Sakurai A, Amornphimoltham P, Masedunskas A, Weigert R, Chavakis T, Adams RH, et al: RhoA and ROCK mediate histamine-induced vascular leakage and anaphylactic shock. Nat Commun. 6(6725)2015.PubMed/NCBI View Article : Google Scholar

90 

Hu X, Yan J, Huang L, Araujo C, Peng J, Gao L, Liu S, Tang J, Zuo G and Zhang JH: INT-777 attenuates NLRP3-ASC inflammasome-mediated neuroinflammation via TGR5/cAMP/PKA signaling pathway after subarachnoid hemorrhage in rats. Brain Behav Immun. 91:587–600. 2021.PubMed/NCBI View Article : Google Scholar

91 

Haselow K, Bode JG, Wammers M, Ehlting C, Keitel V, Kleinebrecht L, Schupp AK, Häussinger D and Graf D: Bile acids PKA-dependently induce a switch of the IL-10/IL-12 ratio and reduce proinflammatory capability of human macrophages. J Leukoc Biol. 94:1253–1264. 2013.PubMed/NCBI View Article : Google Scholar

92 

Kolka CM and Bergman RN: The endothelium in diabetes: Its role in insulin access and diabetic complications. Rev Endocr Metab Disord. 14:13–19. 2013.PubMed/NCBI View Article : Google Scholar

93 

Sampedro J, Bogdanov P, Ramos H, Solà-Adell C, Turch M, Valeri M, Simó-Servat O, Lagunas C, Simó R and Hernández C: New insights into the mechanisms of action of topical administration of GLP-1 in an experimental model of diabetic retinopathy. J Clin Med. 8(339)2019.PubMed/NCBI View Article : Google Scholar

94 

Wang LY, Cheng KC, Li Y, Niu CS, Cheng JT and Niu HS: Glycyrrhizic acid increases glucagon like peptide-1 secretion via TGR5 activation in type 1-like diabetic rats. Biomed Pharmacother. 95:599–604. 2017.PubMed/NCBI View Article : Google Scholar

95 

Claybaugh T, Decker S, McCall K, Slyvka Y, Steimle J, Wood A, Schaefer M, Thuma J and Inman S: L-Arginine supplementation in type II diabetic rats preserves renal function and improves insulin sensitivity by altering the nitric oxide pathway. Int J Endocrinol. 2014(171546)2014.PubMed/NCBI View Article : Google Scholar

96 

Kida T, Tsubosaka Y, Hori M, Ozaki H and Murata T: Bile acid receptor TGR5 agonism induces NO production and reduces monocyte adhesion in vascular endothelial cells. Arterioscler Thromb Vasc Biol. 33:1663–1669. 2013.PubMed/NCBI View Article : Google Scholar

97 

Gloerich M and Bos JL: Epac: Defining a new mechanism for cAMP action. Annu Rev Pharmacol Toxicol. 50:355–375. 2010.PubMed/NCBI View Article : Google Scholar

98 

Lezoualc'h F, Fazal L, Laudette M and Conte C: Cyclic AMP Sensor EPAC proteins and their role in cardiovascular function and disease. Circ Res. 118:881–897. 2016.PubMed/NCBI View Article : Google Scholar

99 

Gündüz D, Troidl C, Tanislav C, Rohrbach S, Hamm C and Aslam M: Role of PI3K/Akt and MEK/ERK Signalling in cAMP/Epac-Mediated endothelial barrier stabilisation. Front Physiol. 10(1387)2019.PubMed/NCBI View Article : Google Scholar

100 

Yuan Y, Engler AJ, Raredon MS, Le A, Baevova P, Yoder MC and Niklason LE: Epac agonist improves barrier function in iPSC-derived endothelial colony forming cells for whole organ tissue engineering. Biomaterials. 200:25–34. 2019.PubMed/NCBI View Article : Google Scholar

101 

Garcia-Morales V, Friedrich J, Jorna LM, Campos-Toimil M, Hammes HP, Schmidt M and Krenning G: The microRNA-7-mediated reduction in EPAC-1 contributes to vascular endothelial permeability and eNOS uncoupling in murine experimental retinopathy. Acta Diabetol. 54:581–591. 2017.PubMed/NCBI View Article : Google Scholar

102 

Ramos CJ, Lin C, Liu X and Antonetti DA: The EPAC-Rap1 pathway prevents and reverses cytokine-induced retinal vascular permeability. J Biol Chem. 293:717–730. 2018.PubMed/NCBI View Article : Google Scholar

103 

Liu L, Jiang Y, Chahine A, Curtiss E and Steinle JJ: Epac1 agonist decreased inflammatory proteins in retinal endothelial cells, and loss of Epac1 increased inflammatory proteins in the retinal vasculature of mice. Mol Vis. 23:1–7. 2017.PubMed/NCBI

104 

Luchetti F, Crinelli R, Cesarini E, Canonico B, Guidi L, Zerbinati C, Di Sario G, Zamai L, Magnani M, Papa S and Iuliano L: Endothelial cells, endoplasmic reticulum stress and oxysterols. Redox Biol. 13:581–587. 2017.PubMed/NCBI View Article : Google Scholar

105 

Song J, Li J, Hou F, Wang X and Liu B: Mangiferin inhibits endoplasmic reticulum stress-associated thioredoxin-interacting protein/NLRP3 inflammasome activation with regulation of AMPK in endothelial cells. Metabolism. 64:428–437. 2015.PubMed/NCBI View Article : Google Scholar

106 

Fiorentino TV, Procopio T, Mancuso E, Arcidiacono GP, Andreozzi F, Arturi F, Sciacqua A, Perticone F, Hribal ML and Sesti G: SRT1720 counteracts glucosamine-induced endoplasmic reticulum stress and endothelial dysfunction. Cardiovasc Res. 107:295–306. 2015.PubMed/NCBI View Article : Google Scholar

107 

Sasaki T, Kuboyama A, Mita M, Murata S, Shimizu M, Inoue J, Mori K and Sato R: The exercise-inducible bile acid receptor Tgr5 improves skeletal muscle function in mice. J Biol Chem. 293:10322–10332. 2018.PubMed/NCBI View Article : Google Scholar

108 

Dicks N, Gutierrez K, Currin L, de Macedo MP, Glanzner WG, Mondadori RG, Michalak M, Agellon LB and Bordignon V: Tauroursodeoxycholic acid/TGR5 signaling promotes survival and early development of glucose-stressed porcine embryos†. Biol Reprod. 105:76–86. 2021.PubMed/NCBI View Article : Google Scholar

109 

Dicks N, Gutierrez K, Currin L, Priotto de Macedo M, Glanzner W, Michalak M, Agellon LB and Bordignon V: Tauroursodeoxycholic acid acts via TGR5 receptor to facilitate DNA damage repair and improve early porcine embryo development. Mol Reprod Dev. 87:161–173. 2020.PubMed/NCBI View Article : Google Scholar

Related Articles

Journal Cover

November-2022
Volume 24 Issue 5

Print ISSN: 1792-0981
Online ISSN:1792-1015

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Zhang M, Dong Z, Dong W, Zhou D and Ren X: Role of Takeda G protein‑coupled receptor 5 in microvascular endothelial cell dysfunction in diabetic retinopathy (Review). Exp Ther Med 24: 674, 2022
APA
Zhang, M., Dong, Z., Dong, W., Zhou, D., & Ren, X. (2022). Role of Takeda G protein‑coupled receptor 5 in microvascular endothelial cell dysfunction in diabetic retinopathy (Review). Experimental and Therapeutic Medicine, 24, 674. https://doi.org/10.3892/etm.2022.11610
MLA
Zhang, M., Dong, Z., Dong, W., Zhou, D., Ren, X."Role of Takeda G protein‑coupled receptor 5 in microvascular endothelial cell dysfunction in diabetic retinopathy (Review)". Experimental and Therapeutic Medicine 24.5 (2022): 674.
Chicago
Zhang, M., Dong, Z., Dong, W., Zhou, D., Ren, X."Role of Takeda G protein‑coupled receptor 5 in microvascular endothelial cell dysfunction in diabetic retinopathy (Review)". Experimental and Therapeutic Medicine 24, no. 5 (2022): 674. https://doi.org/10.3892/etm.2022.11610