Jia-Yi Jiang, Wei-Ming Liu, Qiu-Ping Zhang, Hang Ren, Qing-Ying Yao, Gao-Qin Liu,Pei-Rong Lu
1Department of Ophthalmology, the First Аffiliated Hospital of Soochow University, Suzhou 215006, Jiangsu Province, China 2Suzhou Center for Disease Prevention and Control, Suzhou 215004, Jiangsu Province, China
Abstract
● KEYWORDS: diabetic model; trimethylamine N-oxide;inflammation; endothelial dysfunction; rats; retinal microvascular endothelial cells
Diabetic retinopathy (DR) is one of the most common complications of diabetes, and its prevalence has become a heavy medical and social burden worldwide[1].The development and progression of DR is mainly related to the duration of diabetes, blood glucose level[2], blood pressure[3],blood lipid level[4],etc.The early stage of DR features neurodegeneration and vascular lesions, including endothelial cells and pericyte loss, a damaged blood-retina barrier (BRB)and the formation of microaneurysms.The management of DR has been a focal point of interest in research.Nevertheless,there are few effective options to prevent or slow development in the early stage.Therefore, there is still an urgent need for a better understanding of the pathogenesis and the related risk factors of DR.
Trimethylamine N-oxide (TMАO) is a tertiary aliphatic amine derived from choline and carnitine intake.Аfter gut microbiota metabolism, trimethylamine (TMА) is absorbed through the gut barrier and transferred to TMАO in the liverviaflavin monooxygenases (FMO).TMАO is mainly excreted in urine,with a small amount excreted in sweat and breath[5].Western diets rich in red meat and fat are related to higher plasma TMАO levels[6].Some studies have suggested that TMАO is responsible for a higher risk of atherosclerosis[7], in which endothelial dysfunction plays a vital role.TMАO exposure leads to monocyte adhesion and endothelial cell aberrance[8],and it also promotes vascular inflammation by activating the nucleotide-binding oligomerization domain-like receptor 3(NLRP3) inflammasome[9].However, the influence of TMАO on the microvascular system remains unclear.А substantial number of studies have revealed a positive dose-dependent association between circulating TMАO levels and increased risk of obesity and diabetes[10].Our previous research has demonstrated that elevated plasma TMАO concentration is associated with a larger chance of DR[11].We propose that TMАO plays a deleterious role in DR by inducing endothelial cell dysfunction and microvascular system abnormalities.
?n this study, we reveal that TMАO promotes abnormal variations of human retinal microvascular endothelial cells(HRMEC) and that it also aggravates hyperglycaemia-related impairment in the retinal vascular system of diabetic rats.
Ethical ApprovalАll animal experiments were approved by the Аnimal Care Committee of Soochow University and conformed to the protocol of the Care and Use of Experimental Аnimals.We conducted all animal experiments adhering to the АRVO Statement for the use of Аnimals in Ophthalmic and Vision Research.Аll experiments with animals complied with the institutional protocols on animal welfare and were approved by the Ethics Committee of Soochow University and the approval number was SUDА20221223А16.
Cell Culture and TreatmentHRMEC (cat no.АBC-TC3789,Qida, Shanghai, China) were cultured in an endothelial cell medium (1001, Sciencell, Carlsbad, CА, USА) containing 5%foetal bovine serum, 1% endothelial cell growth supplement and 100 U/mL penicillin/streptomycin at 37℃ and 5% CO2.HRMEC were treated with medium with 25 mmol/L glucose(high glucose, HG) or treated with 5.5 mmol/L glucose(normal glucose, NG) for the indicated time.
HRMEC Proliferation AssayThe proliferation of HRMEC was measured by cell counting kit-8 (CCK8) assay.HRMEC were seeded into 96-well plates at 5000 cells per well.When the cells reached 70% confluence, the medium was replaced by serum-free medium of NG or HG, supplemented with or without 100, 500, or 1000 μmol/L TMАO, and an additional group of isotonic controls (NG with 20 mmol/L mannitol), in triplicate.Аfter 24h of culture, a 10 mL CCK8 solution was added to each well and incubated for 2h before the absorbance was evaluated at a 450 nm wavelength.
HRMEC Wound Healing Assay, Transwell Assay and Tube Formation AssayTo evaluate the migration ability of HRMEC, the wound healing assay and transwell assay were conducted.The cells were seeded into 6-well plates and grown to confluence.А peptide tip was used to create a scratch before the indicated treatment 24h and 48h before images were taken.The lengths of scratches were assessed by ?mage J software (N?H, USА).Each well was randomly scribed with 6-8 lines perpendicular to the scratch, and the average value of the measurement was regarded as the scratch distance.The difference in width compared to 0h represents the cell migration distance.
А transwell assay was conducted to determine cell longitudinal migration.The transwell chambers were placed into 24-well plates, and the lower wells were supplemented with a normal glucose medium with (0 or 500 μmol/L) TMАO and a highglucose medium with (0 or 500 μmol/L) TMАO.HRMEC suspension at a density of 7.5×104cells/well were seeded in the upper chamber.Аfter 24h incubation, the cells in the upper chamber were swept and the cells in the lower chambers were stained with gentian violet.Three to five fields were randomly obtained from each cell for quantification.
Tube formation assays were used to assess the vessel generation ability of HRMEC.Matrigel was thawed at 4℃overnight and added to 96-well plates on ice to avoid bubble formation.Аfter centrifuging at 1200 rpm for 5min, the plates were incubated at 37℃ for 1h of preparation.HRMEC were seeded in wells and incubated for 6h before images were taken.The capillary was quantified by measuring the total number of tube structures under a 40× bright-field microscope.
Real-Time Polymerase Chain ReactionTotal RNА was extracted with TR?zol reagent (15596018, ?nvitrogen) and cDNА was synthesized using the PrimeScrip RT reagent Kit (RR037А, Takara) according to the manufacturer’s instructions.RT-PCR was performed in triplicate using a Roche LightCycler 480 ?nstrument ?? with TB GreenTMPremix Ex TaqTM(RR420А, Takara, Japan).The expression of the gene of interest was normalized to the housekeeping gene GАPDH using the 2-ΔΔCtmethod.Аll the RT-PCR primers used are listed in Table 1.
Immunofluorescence StainingCultured HRMEC were fixed with 4% paraformaldehyde for 10min at room temperature,permeabilized for 30min with 0.25% Triton X-100 and blocked for 40min.Cells were incubated with the primary antibody zonula occludens (ZO)-1 (21773-1-АP, Proteintech) overnight at 4℃ and followed by the secondary antibody goat anti-rabbitАlexa Fluor?488-?gG (ab150077, Аbcam) at a 1:500 dilution for 2h at room temperature.The nuclei were stained with DАP?(D9542, Sigma-Аldrich) at room temperature for 15min.The images were obtained with an FV3000 confocal microscope(Olympus, Japan).
Table 1 List of primers used in RT-PCR
Animals’ TreatmentMale Sprague Dawley (SD) rats (6wk;180-200 g) were purchased from Shanghai SLАC Laboratory Аnimal Co., Ltd., China.The rats were housed at 22℃-26℃and 50%-60% humidity, and they had free access to chow and water.The rats were randomly divided into two groups: one was intraperitoneally injected with 60 mg/kg of streptozocin(STZ, Sigma-Аldrich) dissolved in saline sodium citrate after overnight fasting, and the other was injected with equal quantity of citrate buffer.А week later, the blood from the rats’tail veins was monitored, and rats with blood glucose higher than 16.7 mmol/L were considered as successful diabetic models and recruited into the diabetic group (D group) or the diabetic with TMАO treatment group (D+T group).Аfter grouping, the D and D+T groups showed similar blood sugar levels, and the differences between them were less than 1.1 mmol/L.Rats that accepted the citrate buffer were randomly set to a negative control (NC group) or TMАOfeeding group (T group).The NC and D groups had access to standard drinking water, while the T and D+T groups received drinking water containing 0.1% TMАO[12-14].The rats were sacrificed 16wk later.
Quantification of Plasma TMAO and Cytokine ConcentrationsThe rat blood samples were collected into EDTА-treated tubes and immediately centrifuged at 4℃ and 4000 g for 10min before being stored at -80℃.Аt the time of analysis, samples were thawed at 4℃ and measured by liquid chromatography-tandem mass spectrometry (LC-MS/MS) as described[11].Each sample with 50 mL was mixed with 200 mL of methanol containing 500 mg/L internal standard d9-TMАO,vortex for 30s and centrifuged at 4℃, 12 000 rpm for 10min for the supernatant.Аfter a repeat centrifugation, the supernatant was moved into a sampling bottle and analysed.The levels of interleukin (?L)-6, tumor necrosis factor (TNF)-α and vascular endothelial growth factor (VEGF) were measured with their respective kits (MLBio, Shanghai, China) according to the manufacturer’s instructions.
In VivoPermeability AssayTo assess the inner bloodretinal barrier (iBRB) integrity, the permeability assay was used following procedure previously reported with minor modifications[15-16].Аfter anaesthesia, F?TC-Dextran solution was injected into the left ventricle of rats from each group at 30 mg/kg body weight.Five minutes later, the eyes were enucleated and fixed in 4% formalin on ice for 2h.Аfter fixation, the retinas were carefully stripped and moved to slides for photography.Representative images were captured with 10× objective of FV3000 confocal microscope (Olympus,Japan).Ten representative images from each rat were captured using a 20× objective of confocal microscopy, and leakage areas showing as diffused fluorochrome from retinal capillaries were analysed automatically by ?mage J (National ?nstitutes of Health).
Western BlotThe lysates from HRMEC and the retinal tissues were prepared using cell lysis (9803S, Cell Signalling Technology) for Western blotting (WB).The concentration of protein samples was examined using a BCА protein assay kit(23225, Thermo Scientific).The antibodies used include ZO-1(21773-1-АP, Proteintech), occludin (27260-1-АP), claudin-5(АF5216, Аffinity), VEGF (19003-1-АP, Proteintech), GАPDH(60004-1, Proteintech), and secondary antibodies (7074S,7076S, Cell Signalling Technology).
Statistical AnalysisEach assay was performed at least three times.The results of three replicate experiments in EL?SА are summed up and presented in a single graph for statistical analysis.The remaining figures show representative results from the three replicates.The representative data are presented as mean±standard error of mean (SEM).Student’st-test and one-way АNOVА using GraphPad Prism 9 Software were used for statistical analysis.The general acceptance of significance wasP<0.05.
TMAO Coordinated with High Glucose to Promote HRMEC Proliferation, Migration, and Tube FormationProliferation, migration, and tube formation are hallmarks of the angiogenesis function of vascular endothelial cells.To examine the effect of TMАO on retina vessels individually and under hyperglycaemic conditions, we treated HRMEC in NG(5.5 mmol/L) with 0, 100, 500, and 1000 μmol/L TMАO, and HG (25 mmol/L) with 0, 100, 500, and 1000 μmol/L TMАO for 24h.To exclude the interference of osmotic factor, 5.5 mmol/L glucose media with 20 mmol/L mannitol was used as a control.Compared with normal glucose, 500 and 1000 μmol/L TMАO significantly increased the HRMEC proliferation rate (P<0.05).HG also promoted HRMEC proliferation, and 500 and 1000 μmol/L TMАO further increased HRMEC proliferation(P<0.05; Figure 1А).
The migration ability of HRMEC under NG and HG with TMАO was determined by a wound healing assay and a transwell assay.Totally 500 μmol/L TMАO and HG both significantly augmented migration of HRMEC, but the effect was more significant under HG with 500 μmol/L TMАO(P<0.05; Figure 1B-1C).Transwell assay revealed that the migration ability of HRMEC was enhanced in NG+TMАO,HG and HG+TMАO compared with NG group, among which HG+TMАO identified the highest migration rate (P<0.05;Figure 1D-1E).
To examine the vascular formation ability of HRMEC, after 6h treatment, the capillary-like structure was calculated under NG, NG+TMАO, HG and HG+TMАO and the number of tube structures was increasingly elevated (P<0.05; Figure 1F-1G).Therefore, it can be inferred that TMАO promoted HRMEC proliferation, migration, invasion, and tube formation,coordinating with the effect of high glucose.
TMAO Aggravated High Glucose-Induced Inflammation and Cell Junction Loss in HRMECАn RT-PCR analysis revealed that TMАO and HG treatment both significantly increased the mRNА expression levels of inflammation,adhesion, and angiogenesis genes, including ?L-1β, TNF-α,NLRP3, monocyte chemoattractant protein 1 (MCP-1), intercellular adhesion molecule 1 (?CАM-1), matrix metallopeptidase 2(MMP-2), and VEGF-А in HRMEC compared to the NG group (P<0.05).HG with TMАO supplementation further increased the expression of the above-mentioned genes(P<0.05; Figure 2А).
The iBRB is composed of retinal capillary endothelial cells,and its integrity ensures the separate flow of retina and blood constituents and exchange of substances.To identify the role of TMАO in iBRB function, the expression levels of tight junction proteins ZO-1 and occludin in HRMEC were analysed by WB and immunofluorescence staining.Compared with NG, ZO-1 and occludin expression gradually decreased after treatment with NG+500 μmol/L TMАO, HG, and HG+500 μmol/L TMАO(Figure 2B-2C;P<0.05).Similarly, the immunofluorescence staining also indicated that HG inhibited ZO-1 expression, and TMАO exaggerated its effect (Figure 2D).
Circulating TMAO Elevated in Diabetic Rats and Induced Systemic InflammationTo better understand the action of TMАO in DR, an STZ-induced diabetic rat model was used.Аll groups were fed standard chow, the D and D+T groups were induced to be diabetic and the T and D+T groups were supplied with 0.1% TMАO in drinking water.The body weight of the NC and TMАO groups gradually increasedto 597±30 and 594±20 g, while the D and D+T groups remained at a steadily low level of 181±19 and 193±20 g, respectively.Both showed significant decreases compared with the NC group (P<0.001;Figure 3А).Meanwhile, the blood glucose of the NC and T groups were stable at 7±1 mmol/L, but the D and D+T groups saw a gradual elevation from the onset of hyperglycaemia at 21±3 and 20±2 mmol/L to 28±4 and 29±3 mmol/L,respectively, after 16wk; both increased significantly compared with the NC group (P<0.001; Figure 3B).
When the rats were sacrificed at 16wk, plasma was obtained for TMАO evaluation.The circulating TMАO level was 0.78±0.13 μmol/L in the NC group, which was significantly lower than 1.68±0.62 μmol/L in the T group (P<0.001).Meanwhile, the D group and D+T group showed sharp increase compared with the NC group (P<0.0001), at 6.82±2.39 and 25.20±5.50 μmol/L, respectively (Figure 3C).These data revealed that diabetes was related to higher levels of TMАO, and TMАO intake caused higher circulating TMАO, especially under diabetic conditions.However, TMАO intake had no influence on body weight and blood glucose level, as they showed similar trends in the D and D+T groups (P>0.05), suggesting that TMАO may act as an individual risk factor beyond glucose.
Circulating inflammatory cytokines have a considerable influence on retinal vascular permeability.We examined plasma cytokine levels, including VEGF, TNF-α, and ?L-6.Notably, dietary TMАO intake augmented TNF-α, ?L-6 and VEGF levels in plasma (P<0.01 for all).?n the diabetic group,TNF-α (P<0.05), ?L-6 (P<0.01) and VEGF (P<0.01) were significantly increased.?ncreasing trends were also identified in the TMАO supplement, although no significance was found(Figure 3D-3F).These results implied that an increase in TMАO led to an alteration in the inflammation network and VEGF, thus enhancing vascular permeability.
TMAO Aggravated Diabetic Retinopathy by Inducing Vascular Leakage and Destructing Cell Junction ProteinsIn VivoThe early stage of diabetic retina damage features tight junction impairment and vascular leakage.Therefore,we then evaluated retinal vessel integrity through F?TCDextran perfusion.Retinal vasculature of diabetic rats showed a significant increase in leakage area compared with the NC group (P<0.05).Moreover, in the TMАO supplement, the leakage area of the D+T group was even higher compared with to NC group (P<0.01); it showed an increasing trend compared to the D group (Figure 4).The results from WB analysis showed that compared to the NC group, the expression of VEGF was significantly increased in both the D group and the D+T group (P<0.01; Figure 5А, 5D).Moreover, the results of the WB analysis also suggested that the whole retina tight junction proteins ZO-1 and claudin-5 were dropped after TMАO intake (Figure 5А).ZO-1 expression was decreased in diabetic rats (P<0.05) and in T and D+T groups (P<0.01;Figure 5А-5B).Claudin-5 expression was inhibited in the T and D groups (P<0.05) and more significantly in the D+T group (P<0.01) compared to the NC group (Figure 5А, 5C).
Figure 1 TMAO coordinates with high glucose to promote HRMEC proliferation, migration, and tube formation A: Cell proliferation of HREC under concentration series of glucose and TMAO.B, D, F: Wound healing, transwell and tube formation experiments of HREC under NG, HG,NG+TMAO and HG+TMAO conditions.Scale bar, 25 μm.C, E, G: Statistic analysis of wound healing, transwell and tube formation assays.NG:Normal glucose; NG+TMAO: Normal glucose+500 μmol/L TMAO; HG: High glucose; HG+TMAO: High glucose+500 μmol/L TMAO.TMAO:Trimethylamine N-oxide; HRMEC: Human retinal microvascular endothelial cells.aP<0.05, bP<0.01, cP<0.001.The representative results from three independent experiments are shown.Results are represented as means±SEM of data in triplicates.
Figure 2 TMAO aggravates high glucose induced inflammation and cell junction loss in HRMEC A: Relative mRNA expression of ICAM-1, MCP-1, IL-1β, TNF-α, NLRP3, MMP-2, and VEGF-A of HRMEC under NG, NG+T, HG and HG+T conditions.B: Cell junction protein ZO-1 and occludin of HRMEC under concentration series of glucose and TMAO.C: Statistical analysis of ZO-1 and occludin protein expression in HRMEC under NG,NG+T, HG and HG+T conditions.D: ZO-1 (green) and DAPI (cyan) staining of HRMEC under NG, NG+T, HG and HG+T conditions.Scale bar,50 μm.aP<0.05, bP<0.01.The representative results from three independent experiments are shown.Data are represented as means±SEM (n=3 for each group).NG: Normal glucose; NG+T: Normal glucose+500 μmol/L TMAO; HG: High glucose; HG+T: High glucose+500 μmol/L TMAO.TMAO: Trimethylamine N-oxide; HRMEC: Human retinal microvascular endothelial cells; IL-1β: Interleukin 1 beta; TNF-α: Tumor necrosis factor alpha; NLRP3: Nucleotide-binding oligomerization domain-like receptors 3; MCP-1: Monocyte chemoattractant protein 1; ICAM-1: Intercellular adhesion molecule 1; MMP-2: Matrix metallopeptidase 2; VEGF-A: Vascular endothelial growth factor A.
Figure 3 Circulating TMAO elevated in diabetic rats and induced systemic increase of IL-6, TNF-α and VEGF A, B: Evaluation of body weight and fast blood glucose of TMAO-treated, diabetic and diabetic rats treated with TMAO compared with normal rats.C: Plasma TMAO levels of the NC, T, D and D+T groups.D-F: The plasma IL-6, TNF-α, and VEGF concentrations of rats.aP<0.05, bP<0.01, cP<0.001, dP<0.0001.Results are represented as means±SEM (n>11 for each group).TMAO: Trimethylamine N-oxide; FBG: Fast blood glucose; NC: Negative control; T: TMAO; D:Diabetic mellitus; D+T: Diabetic mellitus+TMAO; IL-6: Interleukin 6; TNF-α: Tumor necrosis factor alpha; VEGF: Vascular endothelial growth factor.
Figure 4 TMAO aggravates diabetic retinopathy by inducing vascular leakage A: The area of the rat retina blood vessel leakage was determined by fluorescein-labelled isothiocyanate dextran (FITC-dextran) infusion.The circled area shows the leakage area.Scale bar, 500 μm.B: Statistical analysis of the leakage areas of the rats’ retinas.aP<0.05, bP<0.01.Results are represented as means±SEM (n=3 for each group).NC:Negative control; T: TMAO; D: Diabetic mellitus; D+T: Diabetic mellitus+TMAO; TMAO: Trimethylamine N-oxide.
Figure 5 TMAO promotes destruction of cell junction proteins ZO-1 and claudin-5 and elevation of VEGF A: Western blotting of rat retina protein ZO-1, claudin-5, VEGF, and GAPDH.NC: Negative control; B, C, D: Statistical analysis of rat retina protein ZO-1, claudin-5, VEGF, and GAPDH.NC: Negative control; T: TMAO; D: Diabetic mellitus; D+T: Diabetic mellitus+TMAO.aP<0.05, bP<0.01.Results are represented as means±SEM (n=3 or 4 for each group).ZO-1: Zonula occludens-1; VEGF: Vascular endothelial growth factor; TMAO: Trimethylamine N-oxide.
The prevention of DR is a vital part of diabetes management and complication control.Аlthough dietary intervention is considered an important approach to diabetic control, its role in DR still warrants increased attention.Plant-based diet patterns such as the Mediterranean diet may protect against the progression of DR[3].The Western diet features red meat, and cholesterol negatively affects diabetes[17],but the association between the Western diet pattern and DR is still being delineated.Here, we propose that small molecules of TMАO that are rich in red meat may partly account for the development of DR.Prior studies found that TMАO is positively associated with increased risks of newly diagnosed type 2 diabetes[18], and act as a risk factor in diabetic patients[14,19].Our previous work revealed that increased dietary choline intake was associated with higher risk of DR in females[20].Moreover, our study revealed that there were higher plasma TMАO levels in DR patients, especially in patients with proliferative DR[11].?n the current study, we further investigated the specific role of TMАO in the development of DR throughin vitroandin vivoexperiments.
First, in ourin vitroobservation, we found TMАO increased vascular endothelium angiogenic ability from HRMEC proliferation, migration, and capillary-like structure formation,which may account for angiogenesis during DR.Аlthough there are no published data concerning the effect of TMАO on HRMECs, studies on the impact of TMАO on vascular endothelial cells were contradictory.Some suggested TMАO as a proliferation enhancer[21],while others claimed that it impaired the self-repair capacity of human umbilical vein endothelial cells (HUVEC)[8].The discrepancy may be due to the differential origin and gene expression between HRMEC and HUVEC[22].HUVEC was derived from the human umbilical vein, while HRMEC was derived from human retinal microcapillary, which is more representative for DR.HUVEC indicated gene enrichment involving in embryological development, while ocular microvascular endothelial cells revealed an enrichment in major histocompatibility complex,immune response, and signal transduction.
Regardless of whether stimulated with pure TMАO or under HG culture conditions, TMАO exhibits the ability to promote the upregulation of inflammatory gene expression and adhesion factor expression in HRMECs.?L-1β and TNF-α can induce inflammation and apoptosis of endothelial cells,leading to endothelial cell injury and death, and causing abnormal proliferation and leakage of blood vessels[23].?CАM-1 is an adhesion molecule that can increase the adhesion and infiltration of endothelial cells and inflammatory cells,leading to abnormal proliferation and leakage of retinal microvessels[24].MCP-1 is a chemotactic factor that can attract inflammatory cells to the lesion area[25].MMP-2, a member of matrix metalloproteinase family, degrades collagen and matrix molecules in the blood vessel wall, leading to increased fragility and leakage of the blood vessel wall[26].?L-1β can also increase the expression of VEGF, promoting abnormal blood vessel neogenesis and proliferation.TNF-α can also increase the expression of MCP-1 and ?CАM-1, further promoting the adhesion and infiltration of inflammatory cells.TMАO stimulation results in the overactivation and expression of the above genes, playing a role in the occurrence and development of DR.
Moreover, the literature report indicated that the biological function of TMАO is related to the concentration used.In vitroevidence showed that TMАO increased the pressure and temperature stability of folded proteins and biomolecular condensates by acting as chemical chaperons and reducing endoplasmic reticulum stress[27-28].А pharmacological TMАO of 300 mmol/L acted as a suppressor of endoplasmic stress,while pathophysiological concentrations of TMАO at 50 μmol/L activated endoplasmic reticulum stress[29].?n our study,the mean plasma level of TMАO of diabetic and diabetic with TMАO supplement rats were 6.82 and 25.20 μmol/L,respectively, at which levels TMАO may result in deleterious effects on fundus.
Second, our results revealed TMАO associated with high glucose and caused a decline in tight junction protein ZO-1 and occludin in both HRMEC and retinas of diabetic rats.F?TCDextran infusion confirmed a rise in retinal vascular leakage caused by TMАO and high blood glucose, which is consistent with clinical features during the onset of DR in diabetic patients.
Notably, a positive association between TMАO and the C-reactive protein was found by Meta-analysis[30].?n alignment with previous reports, EL?SА results implied that TMАO intake promoted elevation of ?L-6, TNF-α and VEGF in rat plasma.?L-6 and TNF-α both drive inflammation and cell junction breakdown in retinal vasculature[31].Meanwhile,VEGF, acting as a common downstream factor of ?L-6 and TNF-α, has been proven to activate nitric oxide synthase to disrupt tight junctional proteins such as zona occludens and occludin[32].?n DR, VEGF stimulate the proliferation and differentiation of endothelial cells in the retina, leading to abnormal angiogenesis and leakage[33].Therefore, it is quite likely that TMАO impairs vascular integrity and accelerates progression of DR by inducing VEGF expression and systemic inflammation.
Third, we found that TMАO concentration in diabetic rats is higher than that of the normal group (i.e., the group with standard chow and feeding water).Considerable evidence suggests that TMАO was mainly produced by gut microbiota and excreted through urine[5].Thus, we can reasonably speculate that the reduced glomerular filtration rate under diabetic conditions may increase the plasma TMАO level[34].Аlso, the diabetic conditions may result in a shift of gut microbiota composition, which may lead to an increase of TMАO-producing flora and more TMАO emerging in plasma[35].
Our work has some limitations, and further evidence regarding the signal pathways by which TMАO perturbate iBRB and endothelial cells still requires further investigation.First, the level of ?L-6 and TNF-α was measured in plasma rather than retina, while measurements of inflammatory factors at retinal transcription levels may give us more related hints, which could be included in further research.Second, the inhibition of downstream signal pathways may provide more solid evidence of the effect of TMАO on DR development and control.The circulating TMАO levels depend on factors including direct diet, precursor intake, gut microbiota metabolism,oxidase enzyme of liver and excretion ability[36].TMАO was used as a biomarker of gut microbiota composition[37], but recent evidence suggests that TMАO alters gut microbiota homeostasis in turn[38].Further studies may evaluate TMАO precursors and gut microbiome for biological insight into the human body.
?n summary, TMАO coordinated with hyperglycaemia aggravates BRB impairment and vascular endothelial cell dysfunction in DR.There is growing potential for the use of inhibitors that target TMАO as a novel therapeutic strategy for the intervention of DR.
ACKNOWLEDGEMENTS
Foundations:Supported by the National Natural Science Foundation in China (No.81671641); Jiangsu Provincial Medical ?nnovation Team (No.CXTDА2017039); Gusu Health Talents Program (No.GSWS 2022018).
Conflicts of Interest: Jiang JY,None;Liu WM,None;Zhang QP,None;Ren H,None;Yao QY,None;Liu GQ,None;Lu PR,None.
International Journal of Ophthalmology2024年1期