Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 8.988
Filter
1.
PLoS One ; 19(9): e0310136, 2024.
Article in English | MEDLINE | ID: mdl-39250437

ABSTRACT

Myocardial fibrosis can trigger heart failure in diabetic cardiomyopathy (DCM), and irisin, an exercise-induced myokine, may have a beneficial effect on cardiac function. However, the specific molecular mechanism between exercise and irisin in the diabetic heart remains not fully explored. This study aimed to investigate how miR-34a mediates exercise-induced irisin to ameliorate myocardial fibrosis and its underlying mechanisms. Type 2 diabetes mellitus (T2DM) with DCM was induced in adult male rats with high-fat diet and streptozotocin injection. The DCM rats were subjected to swimming (60 min/d) and recombinant irisin (r-irisin, 500 µg/kg/d) interventions for 8 weeks, respectively. Cardiac function, cardiomyocyte structure, myocardial fibrosis and its correlated gene and protein expression were analyzed. Swimming intervention alleviated insulin resistance, myocardial fibrosis, and myocardial hypertrophy, and promoted blood glucose homeostasis in T2DM model rats. This improvement was associated with irisin upregulation and miR-34a downregulation in the myocardium, thus enhancing cardiac function. Similar efficacy was observed via intraperitoneal injection of exogenous recombinant irisin. Inhibition of miR-34a in vivo exhibited an anti-myocardial fibrotic effect by promoting irisin secretion through activating sirtuin 1 (SIRT1)/peroxisome proliferator-activated receptor-gamma coactivator-1α (PGC-1α)/fibronectin type III domain-containing protein 5 (FNDC5) signal pathway and downregulating myocardial fibrosis markers (collagen I, collagen III, and transforming growth factor-ß1). Therefore, swimming-induced irisin has the potential therapeutic effect on diabetic myocardial fibrosis through activating the miR-34a-mediated SIRT1/PGC-1α/FNDC5 signal pathway.


Subject(s)
Diabetes Mellitus, Experimental , Diabetes Mellitus, Type 2 , Diabetic Cardiomyopathies , Fibronectins , Fibrosis , MicroRNAs , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Signal Transduction , Sirtuin 1 , Swimming , Animals , Sirtuin 1/metabolism , Sirtuin 1/genetics , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/genetics , Fibronectins/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , Male , Rats , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Experimental/pathology , Diabetic Cardiomyopathies/metabolism , Diabetic Cardiomyopathies/genetics , Diabetic Cardiomyopathies/pathology , Diabetic Cardiomyopathies/etiology , Rats, Sprague-Dawley , Myocardium/metabolism , Myocardium/pathology
2.
Cell Mol Life Sci ; 81(1): 408, 2024 Sep 17.
Article in English | MEDLINE | ID: mdl-39287634

ABSTRACT

Diabetic kidney disease (DKD) is the predominant type of end-stage renal disease. Increasing evidence suggests thatglomerular mesangial cell (MC) inflammation is pivotal for cell proliferation and DKD progression. However, the exactmechanism of MC inflammation remains largely unknown. This study aims to elucidate the role of inflammatoryfactor high-mobility group box 1 (Hmgb1) in DKD. Inflammatory factors related to DKD progression are screened viaRNA sequencing (RNA-seq). In vivo and in vitro experiments, including db/db diabetic mice model, CCK-8 assay, EdUassay, flow cytometric analysis, Co-IP, FISH, qRT-PCR, western blot, single cell nuclear RNA sequencing (snRNA-seq),are performed to investigate the effects of Hmgb1 on the inflammatory behavior of MCs in DKD. Here, wedemonstrate that Hmgb1 is significantly upregulated in renal tissues of DKD mice and mesangial cells cultured withhigh glucose, and Hmgb1 cytopasmic accumulation promotes MC inflammation and proliferation. Mechanistically,Hmgb1 cytopasmic accumulation is two-way regulated by MC-specific cyto-lncRNA E130307A14Rik interaction andlactate-mediated acetylated and lactylated Hmgb1 nucleocytoplasmic translocation, and accelerates NFκB signalingpathway activation via directly binding to IκBα. Together, this work reveals the promoting role of Hmgb1 on MCinflammation and proliferation in DKD and helps expound the regulation of Hmgb1 cytopasmic accumulation in twoways. In particular, Hmgb1 may be a promising therapeutic target for DKD.


Subject(s)
Diabetic Nephropathies , HMGB1 Protein , Mesangial Cells , NF-kappa B , Signal Transduction , HMGB1 Protein/metabolism , HMGB1 Protein/genetics , Animals , Diabetic Nephropathies/metabolism , Diabetic Nephropathies/pathology , Mesangial Cells/metabolism , Mesangial Cells/pathology , Mice , NF-kappa B/metabolism , Male , Cell Proliferation , Disease Progression , Mice, Inbred C57BL , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/pathology , Cytosol/metabolism , Humans , Inflammation/pathology , Inflammation/metabolism
3.
ACS Appl Mater Interfaces ; 16(38): 50160-50174, 2024 Sep 25.
Article in English | MEDLINE | ID: mdl-39265036

ABSTRACT

Diabetic wounds are prone to recurrent infections, often leading to delayed healing. To address this challenge, we developed a chitin-copper sulfide (CuS@CH) composite sponge, which combines bacterial trapping with near-infrared (NIR) activated phototherapy for treating infected diabetic wounds. CuS nanoparticles were synthesized and incorporated in situ within the sponge using a chitin assisted biomineralization strategy. The positively charged chitin surface effectively adhered bacteria, while NIR irradiation of CuS generated reactive oxygen species (ROS) heat and Cu2+ to rapidly damage the trapped bacteria. This synergistic effect resulted in an exceptional antibacterial performance against E. coli (∼99.9%) and S. aureus (∼99.3%). The bactericidal mechanism involved NIR-induced glutathione oxidation, membrane lipid peroxidation, and increased membrane permeability. In diabetic mouse models, the CuS@CH sponge accelerated the wound healing of S. aureus infected wounds by facilitating collagen deposition and reducing inflammation. Furthermore, the sponge demonstrated good biocompatibility. This dual-functional platform integrating bacterial capture and NIR-triggered phototherapy shows promise as an antibacterial wound dressing to promote healing of infected diabetic wound.


Subject(s)
Anti-Bacterial Agents , Chitin , Copper , Diabetes Mellitus, Experimental , Escherichia coli , Infrared Rays , Staphylococcus aureus , Wound Healing , Animals , Wound Healing/drug effects , Staphylococcus aureus/drug effects , Mice , Copper/chemistry , Copper/pharmacology , Escherichia coli/drug effects , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Chitin/chemistry , Chitin/pharmacology , Diabetes Mellitus, Experimental/pathology , Wound Infection/drug therapy , Wound Infection/microbiology , Wound Infection/pathology , Wound Infection/therapy , Reactive Oxygen Species/metabolism , Bandages , Staphylococcal Infections/drug therapy , Staphylococcal Infections/pathology
4.
ACS Appl Mater Interfaces ; 16(38): 50175-50187, 2024 Sep 25.
Article in English | MEDLINE | ID: mdl-39269914

ABSTRACT

Diabetic wound healing including diabetic foot ulcers is a major clinical challenge, which could bring an increased level of mortality and morbidity. However, conventional wound dressings exhibit limited healing efficacy due to their lack of active modulation for the healing process. Here, a near-infrared (NIR) stimuli-responsive composite hydrogel dressing with the synergistic effect of both mechanical contraction and epithelial-mesenchymal transition (EMT) was developed to facilitate cell migration and vascularization for diabetic wound healing. In the methacrylated gelatin-based composite hydrogel, N-isopropylacrylamide and polydopamine nanoparticles were incorporated to endow the composite hydrogel with thermosensitive and photothermal properties. Linagliptin (LIN) was loaded into the composite hydrogel, and the drug release rate could be controlled by NIR laser irradiation. NIR-triggered on-demand active contraction of wound area and LIN release for biological stimulation were potentially realized in this responsive system due to the thermally induced sol-gel transition of the composite hydrogel. The release of loaded LIN could effectively promote cell migration by activating EMT and enhancing angiogenesis. In the full-thickness skin defect model, the LIN-loaded composite hydrogel with NIR laser irradiation had the highest wound closure rate as compared with the pure hydrogel and LIN-loaded hydrogel groups. Therefore, this composite hydrogel can serve as an excellent platform for promoting wound healing and will find more practical value in clinical treatment.


Subject(s)
Cell Movement , Hydrogels , Infrared Rays , Wound Healing , Wound Healing/drug effects , Cell Movement/drug effects , Hydrogels/chemistry , Hydrogels/pharmacology , Animals , Mice , Epithelial-Mesenchymal Transition/drug effects , Epithelial-Mesenchymal Transition/radiation effects , Humans , Polymers/chemistry , Polymers/pharmacology , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Experimental/pathology , Nanoparticles/chemistry , Indoles/chemistry , Indoles/pharmacology
5.
Physiol Rep ; 12(18): e70016, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39294856

ABSTRACT

The carotid body (CB) senses changes in arterial O2 partial pressure (pO2) and glucose levels; therefore, it is key for the detection of hypoxia and hypoglycemia. The CB has been suggested to detect pO2 through an increase in reactive oxygen species (ROS) in the mitochondria. However, the mechanism protecting the chemoreceptor cells and their mitochondria from ROS and hyperglycemia is poorly understood. Here we measured glutathione levels in CB mitochondria of control and in streptozotocin (STZ)-induced type 1 diabetic male Wistar rats. We found a dramatic reduction in total glutathione from 11.45 ± 1.30 µmol/mg protein in control rats to 1.45 ± 0.31 µmol/mg protein in diabetic rats. However, the ratio of reduced to oxidized glutathione, a measure of the redox index, was increased in diabetic rats compared to controls. We conclude that the mitochondria of CB chemoreceptor cells in type 1 diabetic male Wistar rats were likely under glutathione-reducing stress.


Subject(s)
Carotid Body , Diabetes Mellitus, Experimental , Glutathione , Mitochondria , Rats, Wistar , Animals , Male , Carotid Body/metabolism , Rats , Mitochondria/metabolism , Glutathione/metabolism , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/pathology , Diabetes Mellitus, Type 1/metabolism , Oxidative Stress , Reactive Oxygen Species/metabolism , Oxidation-Reduction
6.
Int J Med Sci ; 21(12): 2365-2378, 2024.
Article in English | MEDLINE | ID: mdl-39310260

ABSTRACT

Enhancement of Connexin43 (Cx43) and ferroptosis are respectively associated with the exacerbation of myocardial ischemia-reperfusion injury (MIRI) in diabetes. Myocardial vulnerability to ischemic insult has been shown to vary during early and later phases of diabetes in experimental settings. Whether or not Connexin43 (Cx43) and ferroptosis interplay during MIRI in diabetes is unknown. We, thus, aimed to investigate whether or not the content of myocardial Cx43 may be attributable to myocardial vulnerability to MIRI at different stages of diabetes and also to explore the potential interplay between Cx43 and ferroptosis in this pathology. Age-matched control and subgroups of Streptozotocin-induced diabetic mice were subjected to MIRI induced by 30 minutes coronary artery occlusion and 2 hours reperfusion respectively at 1, 2 and 5 weeks of diabetes. Rat cardiac H9C2 cells were exposed to high glucose (HG) for 48h in the absence or presence of Cx43 gene knockdown followed by hypoxia/reoxygenation (HR) respectively for 6 and 12 hours. Post-ischemic myocardial infarct size was reduced in 1 and 2 weeks DM mice concomitant with enhanced GPX4 and reduced cardiac Cx43 and ferroptosis as compared to control. By contrast, cardiac GPX4 was significantly reduced while Cx43 increased at DM 5 weeks (D5w) which was correspondent to significant increases in ferroptosis and myocardial infarction. Post-ischemic cardiac function was improved in 1 and 2 weeks but worsened in 5w DM mice as compared with non-diabetic control. GAP19 (Cx43 inhibitor) significantly attenuated ferroptosis and reduced myocardial infarction in D5w mice. Erastin (ferroptosis activator) reversed the cardioprotective effect of GAP19. In vitro, HR significantly reduced cell viability accompanied with reduced GPX4 but elevated Cx43 expression, MDA production and ferroptosis. Cx43 gene knockdown in H9C2 resulted in a significant increase in GPX4, reduction in MDA and ferroptosis, and subsequently reduced post-hypoxic cell viability. The beneficial effects of Cx43 gene knock-down was minified or eliminated by Erastin. It is concluded that Cx43 overexpression exacerbates MIRI under diabetic conditions via promoting ferroptosis, while its down-regulation at early state of diabetes is attributable to enhanced myocardial tolerance to MIRI.


Subject(s)
Connexin 43 , Diabetes Mellitus, Experimental , Diabetes Mellitus, Type 1 , Ferroptosis , Myocardial Reperfusion Injury , Phospholipid Hydroperoxide Glutathione Peroxidase , Animals , Ferroptosis/genetics , Connexin 43/metabolism , Connexin 43/genetics , Mice , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/pathology , Myocardial Reperfusion Injury/genetics , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Experimental/pathology , Rats , Phospholipid Hydroperoxide Glutathione Peroxidase/metabolism , Phospholipid Hydroperoxide Glutathione Peroxidase/genetics , Diabetes Mellitus, Type 1/complications , Diabetes Mellitus, Type 1/pathology , Diabetes Mellitus, Type 1/metabolism , Diabetes Mellitus, Type 1/genetics , Male , Gene Knockdown Techniques , Humans , Cell Line , Myocardium/pathology , Myocardium/metabolism , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Myocardial Infarction/pathology , Myocardial Infarction/genetics , Myocardial Infarction/metabolism
7.
J Pineal Res ; 76(6): e13008, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39300782

ABSTRACT

Diabetic retinopathy (DR) is characterized as a microvascular disease. Nonproliferative diabetic retinopathy (NPDR) presents with alterations in retinal blood flow and vascular permeability, thickening of the basement membrane, loss of pericytes, and formation of acellular capillaries. Endothelial-mesenchymal transition (EndMT) of retinal microvessels may play a critical role in advancing NPDR. Melatonin, a hormone primarily secreted by the pineal gland, is a promising therapeutic for DR. This study explored the EndMT in retinal microvessels of NPDR and its related mechanisms. The effect of melatonin on the retina of diabetic rats was evaluated by electroretinogram (ERG) and histopathologic slide staining. Furthermore, the effect of melatonin on human retinal microvascular endothelial cells (HRMECs) was detected by EdU incorporation assay, scratch assay, transwell assay, and tube formation test. Techniques such as RNA-sequencing, overexpression or knockdown of target genes, extraction of cytoplasmic and nuclear protein, co-immunoprecipitation (co-IP), and multiplex immunofluorescence facilitated the exploration of the mechanisms involved. Our findings reveal, for the first time, that melatonin attenuates diabetic retinopathy by regulating EndMT of retinal vascular endothelial cells via inhibiting the HDAC7/FOXO1/ZEB1 axis. Collectively, these results suggest that melatonin holds potential as a therapeutic strategy to reduce retinal vascular damage and protect vision in NPDR.


Subject(s)
Diabetes Mellitus, Experimental , Diabetic Retinopathy , Endothelial Cells , Histone Deacetylases , Melatonin , Zinc Finger E-box-Binding Homeobox 1 , Melatonin/pharmacology , Diabetic Retinopathy/metabolism , Diabetic Retinopathy/drug therapy , Diabetic Retinopathy/pathology , Animals , Rats , Endothelial Cells/metabolism , Endothelial Cells/drug effects , Histone Deacetylases/metabolism , Zinc Finger E-box-Binding Homeobox 1/metabolism , Zinc Finger E-box-Binding Homeobox 1/genetics , Humans , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/pathology , Male , Forkhead Box Protein O1/metabolism , Retinal Vessels/drug effects , Retinal Vessels/metabolism , Retinal Vessels/pathology , Rats, Sprague-Dawley , Epithelial-Mesenchymal Transition/drug effects , Retina/metabolism , Retina/drug effects , Retina/pathology , Endothelial-Mesenchymal Transition
8.
J Cell Mol Med ; 28(17): e70035, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39245790

ABSTRACT

Diabetes-related bone loss represents a significant complication that persistently jeopardizes the bone health of individuals with diabetes. Primary cilia proteins have been reported to play a vital role in regulating osteoblast differentiation in diabetes-related bone loss. However, the specific contribution of KIAA0753, a primary cilia protein, in bone loss induced by diabetes remains unclear. In this investigation, we elucidated the pivotal role of KIAA0753 as a promoter of osteoblast differentiation in diabetes. RNA sequencing demonstrated a marked downregulation of KIAA0753 expression in pro-bone MC3T3 cells exposed to a high glucose environment. Diabetes mouse models further validated the downregulation of KIAA0753 protein in the femur. Diabetes was observed to inhibit osteoblast differentiation in vitro, evidenced by downregulating the protein expression of OCN, OPN and ALP, decreasing primary cilia biosynthesis, and suppressing the Hedgehog signalling pathway. Knocking down KIAA0753 using shRNA methods was found to shorten primary cilia. Conversely, overexpression KIAA0753 rescued these changes. Additional insights indicated that KIAA0753 effectively restored osteoblast differentiation by directly interacting with SHH, OCN and Gli2, thereby activating the Hedgehog signalling pathway and mitigating the ubiquitination of Gli2 in diabetes. In summary, we report a negative regulatory relationship between KIAA0753 and diabetes-related bone loss. The clarification of KIAA0753's role offers valuable insights into the intricate mechanisms underlying diabetic bone complications.


Subject(s)
Cell Differentiation , Microtubule-Associated Proteins , Osteoblasts , Signal Transduction , Animals , Humans , Male , Mice , Cell Line , Cilia/metabolism , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/pathology , Diabetes Mellitus, Experimental/genetics , Hedgehog Proteins/metabolism , Hedgehog Proteins/genetics , Mice, Inbred C57BL , Osteoblasts/metabolism , Osteogenesis/genetics , Microtubule-Associated Proteins/metabolism
9.
Ann Clin Lab Sci ; 54(4): 539-546, 2024 Jul.
Article in English | MEDLINE | ID: mdl-39293843

ABSTRACT

OBJECTIVE: To study the effect of dexmedetomidine (Dex) on myocardial injury induced by acute kidney injury (AKI) in diabetes mellitus rats and explore the potential mechanisms. METHODS: The Type 2 diabetes mellitus (T2DM) model was prepared in 40 adult male Wistar rats. These rats were randomly divided into four groups (n=10/group), including the control (Con) group, AKI group, Dex preconditioning (DPreC) group, and resveratrol (Res) combined with Dex preconditioning (Res+DPreC) group. The AKI model was prepared in the AKI, DPreC, and Res+DPreC group. The DPreC group received Dex, while the Con and AKI group received normal saline. The Res+DPreC group received Res in addition to Dex preconditioning. Histopathologic, apoptotic, enzymatic, and inflammatory changes in myocardial tissue were observed or detected. RESULTS: Histopathologic, apoptotic, and enzymatic changes in myocardial tissue demonstrated that AKI induced myocardial injury in T2DM rats; Dex preconditioning could mitigate this injury; and RES enhanced this effect. Inflammatory changes suggested that Dex alleviated the inflammatory response induced by AKI in T2DM rats via regulating the expressions of SIRT1, TNF-α, IL-17A, and IL-10. CONCLUSIONS: Dex could alleviate myocardial injury induced by AKI in DM rats via regulating the inflammatory response associated with SIRT1, TNF-α, IL-17A, and IL-10, and Res could enhance this protective effect.


Subject(s)
Acute Kidney Injury , Dexmedetomidine , Diabetes Mellitus, Experimental , Inflammation , Rats, Wistar , Animals , Dexmedetomidine/pharmacology , Dexmedetomidine/therapeutic use , Acute Kidney Injury/drug therapy , Acute Kidney Injury/etiology , Acute Kidney Injury/pathology , Acute Kidney Injury/metabolism , Male , Rats , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Experimental/pathology , Inflammation/pathology , Inflammation/drug therapy , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/drug therapy , Myocardium/pathology , Myocardium/metabolism , Resveratrol/pharmacology , Sirtuin 1/metabolism , Apoptosis/drug effects
10.
Sci Rep ; 14(1): 20633, 2024 09 04.
Article in English | MEDLINE | ID: mdl-39232184

ABSTRACT

Herbs have been used as medicines since antiquity, and it has been discovered that the human body responds well to herbal remedies. Research on the effect of butin was conducted in the current study in the alloxan-induced diabetic rat paradigm. A total of 30 Wistar rats were randomly assigned into the following groups (n = 6): I-Normal; II-Alloxan-induced (50 mg/kg); III-Alloxan + butin 25 mg/kg; IV-Alloxan + butin 50 mg/kg; V-Butin per se 50 mg/kg. Various diabetic parameters (blood glucose, insulin, HbA1c), lipid profile, inflammatory (TNF-α, IL-1ß, IL-6 and NF-κB), antioxidant enzymes (CAT, SOD and GSH), oxidative stress indicators (MDA), apoptosis marker (caspase-3), hepatic markers (ALT and AST), and histopathological changes were assessed. Additionally, molecular docking and dynamics were performed to evaluate the interaction of butin with target proteins. Butin treatment, at both doses, significantly restored biochemical parameters and preserved pancreatic histopathology in diabetic rats. It effectively modulated blood parameters, lipid profiles, inflammatory markers, apoptosis, antioxidant enzyme activity, oxidative stress, and hepatic markers. Molecular docking revealed that butin binds to proteins such as caspase-3 (1NME), NF-κB (1SVC), and serum insulin (4IBM) with binding affinities of - 7.4, - 6.5, and - 8.2 kcal/mol, respectively. Molecular dynamics simulations further suggested that butin induces significant conformational changes in these proteins. Butin exhibits potential effects against alloxan-induced diabetic rats by restoring biochemical balance, reducing inflammation, and protecting pancreatic tissue. Its binding to key proteins involved in apoptosis and inflammation highlights its therapeutic potential in diabetes management.


Subject(s)
Alloxan , Diabetes Mellitus, Experimental , Molecular Docking Simulation , Rats, Wistar , Animals , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/pathology , Rats , Male , Oxidative Stress/drug effects , Antioxidants/pharmacology , Antioxidants/metabolism , Blood Glucose/metabolism , Pancreas/pathology , Pancreas/drug effects , Pancreas/metabolism , Apoptosis/drug effects , Insulin/metabolism , Hypoglycemic Agents/pharmacology , Hypoglycemic Agents/chemistry , Liver/metabolism , Liver/drug effects , Liver/pathology , Molecular Dynamics Simulation
11.
Sci Rep ; 14(1): 20136, 2024 08 29.
Article in English | MEDLINE | ID: mdl-39209899

ABSTRACT

Pregestational diabetes mellitus (PGDM) has an impact on fetal bone formation, but the underlying mechanism is still obscure. Although miRNAs have been extensively investigated throughout bone formation, their effects on fetal bone development caused by PGDM still need clarification. This study intends to examine the mechanism by which hyperglycemia impairs the bone formation of offspring via miR-322-5p (miR-322). In this study, miR-322 was selected by systemically screening utilizing bioinformatics and subsequent validation experiments. Using streptozotocin (STZ)-induced diabetic mice and ATDC5 cell lines, we found that miR-322 was abundantly expressed in the proliferative and hypertrophic zones of the growth plate, and its expression pattern was disturbed in the presence of hyperglycemia, suggesting that miR-322 is involved in the chondrocyte proliferation and differentiation in absence/presence of hyperglycemia. This observation was proved by manipulating miR-322 expression in ATDC5 cells by transfecting mimic and inhibitor of miR-322. Furthermore, Adamts5, Col12a1, and Cbx6 were identified as the potential target genes of miR-322, verified by the co-transfection of miR-322 inhibitor and the siRNAs, respectively. The evaluation criteria are the chondrocyte proliferation and differentiation and their relevant key gene expressions (proliferation: Sox9 and PthIh; differentiation: Runx2 and Col10a1) after manipulating the gene expressions in ATDC5 cells. This study revealed the regulative role miR-322 on chondrocyte proliferation and differentiation of growth plate by targeting Adamts5, Col12a1, and Cbx6 in hyperglycemia during pregnancy. This translational potential represents a promising avenue for advancing our understanding of bone-related complications in diabetic pregnancy and mitigating bone deficiencies in diabetic pregnant individuals, improving maternal and fetal outcomes.


Subject(s)
Cell Differentiation , Cell Proliferation , Chondrocytes , Diabetes Mellitus, Experimental , Diabetes, Gestational , Growth Plate , MicroRNAs , Animals , Female , Mice , Pregnancy , Cell Differentiation/genetics , Cell Line , Cell Proliferation/genetics , Chondrocytes/metabolism , Chondrocytes/pathology , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/pathology , Diabetes, Gestational/metabolism , Diabetes, Gestational/genetics , Diabetes, Gestational/pathology , Growth Plate/metabolism , Growth Plate/pathology , MicroRNAs/genetics , MicroRNAs/metabolism
12.
Acta Biomater ; 186: 454-469, 2024 Sep 15.
Article in English | MEDLINE | ID: mdl-39098446

ABSTRACT

Diabetic wound treatment continues to be a significant clinical issue due to higher levels of oxidative stress, susceptibility to bacterial infections, and chronic inflammatory responses during healing. We rationally developed and synthesized an ultra-small carbon dots (C-dots) loaded with zinc single-atom nanozyme (Zn/C-dots) with the aim of promoting wounds healing by nanocatalytic treatment, especially targeting its complex pathological microenvironment. Zinc single atoms and C-dots form a dual catalytic system with higher enzymatic activity. Furthermore, the Zn/C-dots nanozyme effectively enters cells, accumulates at mitochondria, and removes excess ROS, protecting cells from oxidative stress damage and limiting the release of pro-inflammatory cytokines, hence reducing inflammation. Zinc can synergistically increase the antibacterial action of C-dots (the effective antibacterial rate of 100 µg/mL Zn/C-dots was above 90 %). Unlike traditional C-dots, Zn/C-dots can cause endothelial cell migration and the formation of new blood vessels. In vitro cytotoxicity, blood compatibility, and in vivo toxicity studies of Zn/C-dots show that they are biocompatible. We subsequently utilized the Zn/C-dots nanozymes to treat diabetic rats' chronic wounds for external use, combining them with ROS-responsive hydrogels to create an antioxidative system (H-Zn/C-dots). The hydrogels anchored the Zn/C-dots nanozymes to the wound, allowing for long-term treatment. The results revealed that H-Zn/C-dots can considerably reduce inflammation, accelerate angiogenesis, collagen deposition, and promote tissue remodeling at the diabetic wound site. After 14 days, the wound area had decreased to approximately 9.19 %, making it a potential treatment. STATEMENT OF SIGNIFICANCE: An ultra-small carbon dot with a zinc single-atom nanozyme was designed and manufactured. Zn/C-dots possess antibacterial, ROS-scavenging, and angiogenesis activities. In vivo, the multifunctional ROS-responsive hydrogel incorporating Zn/C-dots could speed up diabetic wound healing.


Subject(s)
Carbon , Diabetes Mellitus, Experimental , Wound Healing , Zinc , Animals , Zinc/chemistry , Zinc/pharmacology , Wound Healing/drug effects , Carbon/chemistry , Carbon/pharmacology , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Experimental/pathology , Humans , Catalysis , Male , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Rats , Rats, Sprague-Dawley , Quantum Dots/chemistry , Quantum Dots/therapeutic use , Human Umbilical Vein Endothelial Cells , Hydrogels/chemistry , Hydrogels/pharmacology , Reactive Oxygen Species/metabolism
13.
Life Sci Alliance ; 7(10)2024 Oct.
Article in English | MEDLINE | ID: mdl-39117458

ABSTRACT

Neuroinflammation, aging, and neurodegenerative disorders are associated with excessive accumulation of neutral lipids in lipid droplets (LDs) in microglia. Type 2 diabetes mellitus (T2DM) may cause neuroinflammation and is a risk factor for neurodegenerative disorders. Here, we show that hippocampal pyramidal neurons contain smaller, more abundant LDs than their neighboring microglia. The density of LDs varied between pyramidal cells in adjacent subregions, with CA3 neurons containing more LDs than CA1 neurons. Within the CA3 region, a gradual increase in the LD content along the pyramidal layer from the hilus toward CA2 was observed. Interestingly, the high neuronal LD content correlated with less ramified microglial morphotypes. Using the db/db model of T2DM, we demonstrated that diabetes increased the number of LDs per microglial cell without affecting the neuronal LD density. High-intensity interval exercise induced smaller changes in the number of LDs in microglia but was not sufficient to counteract the diabetes-induced changes in LD accumulation. The changes observed in response to T2DM may contribute to the cerebral effects of T2DM and provide a mechanistic link between T2DM and neurodegenerative disorders.


Subject(s)
Diabetes Mellitus, Type 2 , Hippocampus , Lipid Droplets , Microglia , Neurons , Microglia/metabolism , Animals , Lipid Droplets/metabolism , Hippocampus/metabolism , Hippocampus/pathology , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/pathology , Neurons/metabolism , Neurons/pathology , Male , Mice , Physical Conditioning, Animal , Pyramidal Cells/metabolism , Pyramidal Cells/pathology , Mice, Inbred C57BL , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/pathology , Lipid Metabolism , Neuroinflammatory Diseases/metabolism , Neuroinflammatory Diseases/pathology
14.
Redox Rep ; 29(1): 2382943, 2024 Dec.
Article in English | MEDLINE | ID: mdl-39092597

ABSTRACT

OBJECTIVES: Diabetes is closely linked to hearing loss, yet the exact mechanisms remain unclear. Cochlear stria vascularis and pericytes (PCs) are crucial for hearing. This study investigates whether high glucose induces apoptosis in the cochlear stria vascularis and pericytes via elevated ROS levels due to oxidative stress, impacting hearing loss. METHODS: We established a type II diabetes model in C57BL/6J mice and used auditory brainstem response (ABR), Evans blue staining, HE staining, immunohistochemistry, and immunofluorescence to observe changes in hearing, blood-labyrinth barrier (BLB) permeability, stria vascularis morphology, and apoptosis protein expression. Primary cultured stria vascularis pericytes were subjected to high glucose, and apoptosis levels were assessed using flow cytometry, Annexin V-FITC, Hoechst 33342 staining, Western blot, Mitosox, and JC-1 probes. RESULTS: Diabetic mice showed decreased hearing thresholds, reduced stria vascularis density, increased oxidative stress, cell apoptosis, and decreased antioxidant levels. High glucose exposure increased apoptosis and ROS content in pericytes, while mitochondrial membrane potential decreased, with AIF and cytochrome C (CytC) released from mitochondria to the cytoplasm. Adding oxidative scavengers reduced AIF and CytC release, decreasing pericyte apoptosis. DISCUSSION: Hyperglycemia may induce mitochondrial apoptosis of cochlear stria vascularis pericytes through oxidative stress.


Subject(s)
Apoptosis Inducing Factor , Apoptosis , Cytochromes c , Hyperglycemia , Mice, Inbred C57BL , Mitochondria , Oxidative Stress , Pericytes , Proto-Oncogene Proteins c-bcl-2 , Reactive Oxygen Species , Stria Vascularis , Animals , Pericytes/metabolism , Pericytes/drug effects , Pericytes/pathology , Stria Vascularis/metabolism , Stria Vascularis/pathology , Mice , Reactive Oxygen Species/metabolism , Mitochondria/metabolism , Cytochromes c/metabolism , Apoptosis Inducing Factor/metabolism , Hyperglycemia/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Male , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/pathology , Cochlea/metabolism , Cochlea/pathology
15.
ACS Nano ; 18(33): 21998-22009, 2024 Aug 20.
Article in English | MEDLINE | ID: mdl-39115238

ABSTRACT

Lipid droplets (LDs), the essential cytosolic fat storage organelles, have emerged as pivotal regulators of cellular metabolism and are implicated in various diseases. The noninvasive monitoring of LDs necessitates fluorescent probes with precise organelle selectivity and biocompatibility. Addressing this need, we have engineered a probe by strategically modifying the structure of a conventional two-photon-absorbing dipolar dye, acedan. This innovative approach induces nanoaggregate formation in aqueous environments, leading to aggregation-induced fluorescence quenching. Upon cellular uptake via clathrin-mediated endocytosis, the probe selectively illuminates within LDs through a disassembly process, effectively distinguishing LDs from the cytosol with exceptional specificity. This breakthrough enables the high-fidelity imaging of LDs in both cellular and tissue environments. In a pioneering investigation, we probed LDs in a diabetes model induced by streptozotocin, unveiling significantly heightened LD accumulation in cardiac tissues compared to other organs, as evidenced by TP imaging. Furthermore, our exploration of a lipopolysaccharide-mediated cardiomyopathy model revealed an LD accumulation during heart injury. Thus, our developed probe holds immense potential for elucidating LD-associated diseases and advancing related research endeavors.


Subject(s)
Clathrin , Fluorescent Dyes , Lipid Droplets , Animals , Lipid Droplets/metabolism , Lipid Droplets/chemistry , Clathrin/metabolism , Fluorescent Dyes/chemistry , Mice , Endocytosis , Diabetes Mellitus, Experimental/chemically induced , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/pathology , Diabetes Mellitus, Experimental/diagnostic imaging , Photons , Humans , Optical Imaging , Male , Mice, Inbred C57BL
16.
J Biophotonics ; 17(8): e202400115, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39155125

ABSTRACT

Vision impairment caused by diabetic retinopathy (DR) is often irreversible, making early-stage diagnosis imperative. Raman spectroscopy emerges as a powerful tool, capable of providing molecular fingerprints of tissues. This study employs RS to detect ex vivo retinal tissue from diabetic rats at various stages of the disease. Transmission electron microscopy was utilized to reveal the ultrastructural changes in retinal tissue. Following spectral preprocessing of the acquired data, the random forest and orthogonal partial least squares-discriminant analysis algorithms were employed for spectral data analysis. The entirety of Raman spectra and all annotated bands accurately and distinctly differentiate all animal groups, and can identify significant molecules from the spectral data. Bands at 524, 1335, 543, and 435 cm-1 were found to be associated with the preproliferative phase of DR. Bands at 1045 and 1335 cm-1 were found to be associated with early stages of DR.


Subject(s)
Diabetic Retinopathy , Machine Learning , Spectrum Analysis, Raman , Animals , Diabetic Retinopathy/pathology , Rats , Male , Diabetes Mellitus, Experimental/pathology , Diabetes Mellitus, Experimental/chemically induced , Streptozocin , Retina/pathology , Retina/diagnostic imaging , Rats, Sprague-Dawley
17.
J Physiol Investig ; 67(4): 215-224, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-39206781

ABSTRACT

ABSTRACT: Diabetic retinopathy (DR) is one of the significant disabling outcomes of diabetes mellitus characterized by retinal microvascular damage, inflammation, and neuronal dysfunction. Allicin (Alc), a natural compound found in garlic, has garnered attention for its antioxidant and anti-inflammatory properties, positioning it as a potential therapeutic agent for DR. The aim of the present study was to investigate the therapeutic efficacy of Alc in DR management and elucidate its underlying mechanisms of action. We established a DR model in male Sprague-Dawley rats (n = 50, 200-250 g, 12 weeks old) using a high-fat diet for 8 weeks plus a low dose of streptozotocin administered at the start of the 4th week. The diabetic (Diab) animals were administered Alc (16 mg/kg/day, orally), either alone or in combination with mitochondrial division inhibitor-1 (Mdivi-1) as a mitophagy inhibitor, starting 28 days before tissue sampling. We evaluated histopathological changes, metabolic abnormalities associated with type 2 diabetes mellitus (T2DM), the expression of proteins regulating pyroptosis (NOD-like receptor family pyrin domain containing 3, cleaved-caspase 1, and gasdermin D-N terminal) and mitophagy (phosphatase and tensin homolog-induced kinase 1 [PINK1] and Parkin), as well as the levels of oxidative stress mediators and proinflammatory cytokines. Alc treatment effectively ameliorated histopathological changes and metabolic abnormalities associated with T2DM. It downregulated pyroptosis-related proteins, upregulated mitophagy-related proteins, reduced proinflammatory cytokine levels, and attenuated oxidative stress. Treatment with Mdivi-1 suppressed the beneficial effects of Alc. Our findings highlight the therapeutic potential of Alc in managing DR by targeting multiple pathophysiological pathways, including pyroptosis, inflammation, and oxidative stress. The observed antipyroptotic effects of Alc were partially mediated by the activation of the PINK1/parkin-mediated mitophagy pathway. Additional studies are necessary to thoroughly understand the therapeutic mechanisms of Alc and its viability as a treatment choice for DR.


Subject(s)
Diabetes Mellitus, Experimental , Diabetic Retinopathy , Disulfides , Inflammasomes , Mitophagy , Oxidative Stress , Rats, Sprague-Dawley , Sulfinic Acids , Ubiquitin-Protein Ligases , Animals , Male , Oxidative Stress/drug effects , Mitophagy/drug effects , Rats , Sulfinic Acids/pharmacology , Sulfinic Acids/therapeutic use , Inflammasomes/drug effects , Inflammasomes/metabolism , Diabetic Retinopathy/drug therapy , Diabetic Retinopathy/pathology , Diabetic Retinopathy/metabolism , Disulfides/pharmacology , Disulfides/therapeutic use , Ubiquitin-Protein Ligases/metabolism , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Experimental/pathology , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Protein Kinases/metabolism
18.
Sci Rep ; 14(1): 20251, 2024 08 31.
Article in English | MEDLINE | ID: mdl-39215017

ABSTRACT

Monosodium urate (MSU) crystallisation deposited in local tissues and organs induce inflammatory reactions, resulting in diseases such as gout. MSU has been recognized as a common and prevalent pathology in various clinical conditions. In this study, we investigated the role of MSU in the pathogenesis of diabetic kidney disease (DKD). We induced renal injury in diabetic kidney disease mice using streptozotocin (STZ) and assessed renal histopathological damage using Masson's trichrome staining and Collagen III immunofluorescence staining. We measured the levels of malondialdehyde (MDA), superoxide dismutase (SOD), and uric acid (UA) using ELISA. Protein expression levels of NLRP3, p-NF-κB, SHP2, p-STAT3, and p-ERK1/2 were analyzed by Western blot. To further investigate the role of MSU in diabetic kidney disease, we conducted in vitro experiments. In our in vivo experiments, we found that compared to the Model group, there was a significant increase in interstitial fibrosis in the kidneys of mice after treatment with MSU, accompanied by elevated levels of MDA, SOD, and UA. Furthermore, the protein expression of NLRP3, p-NF-NB, SHP2, p-STAT3, and p-ERK1/2 was upregulated. In our subsequent studies on mouse fibroblasts (L929 cells), we discovered that high glucose, MSU, and TGF-ß could promote the expression of P22, GP91, NLRP3, NF-κB, p-NF-κB, p-SHP2, p-EGFR, p-STAT3, and Collagen-III proteins. Additionally, we found that SHP2 could counteract the upregulation trend induced by MSU on the expression of p-SHP2, p-EGFR, p-STAT3, and Collagen-III proteins, and inhibitors YQ128, NAC, and Cetuximab exhibited similar effects. Furthermore, immunofluorescence results indicated that SHP2 could inhibit the expression of the fibrosis marker α-SMA in L929 cells. These findings suggest that MSU can promote renal fibroblast SHP2 expression, induce oxidative stress, activate the NLRP3/NF-κB pathway, and enhance diabetic kidney disease fibroblast proliferation through the TGFß/STAT3/ERK1/2 signaling pathway, leading to renal fibrosis.


Subject(s)
Cell Proliferation , Diabetic Nephropathies , Fibroblasts , Fibrosis , Signal Transduction , Transforming Growth Factor beta , Animals , Male , Mice , Crystallization , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Experimental/pathology , Diabetic Nephropathies/metabolism , Diabetic Nephropathies/pathology , Fibroblasts/metabolism , Fibroblasts/pathology , Fibrosis/metabolism , Kidney/metabolism , Kidney/pathology , Mice, Inbred C57BL , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 11/metabolism , Reactive Oxygen Species/metabolism , STAT3 Transcription Factor/metabolism , Transforming Growth Factor beta/metabolism , Uric Acid/metabolism
19.
Cell Rep Med ; 5(9): 101694, 2024 Sep 17.
Article in English | MEDLINE | ID: mdl-39173634

ABSTRACT

Patients with diabetes often experience fragile fractures despite normal or higher bone mineral density (BMD), a phenomenon termed the diabetic bone paradox (DBP). The pathogenesis and therapeutics opinions for diabetic bone disease (DBD) are not fully explored. In this study, we utilize two preclinical diabetic models, the leptin receptor-deficient db/db mice (DB) mouse model and the streptozotocin-induced diabetes (STZ) mouse model. These models demonstrate higher BMD and lower mechanical strength, mirroring clinical observations in diabetic patients. Advanced glycation end products (AGEs) accumulate in diabetic bones, causing higher non-enzymatic crosslinking within collagen fibrils. This inhibits intrafibrillar mineralization and leads to disordered mineral deposition on collagen fibrils, ultimately reducing bone strength. Guanidines, inhibiting AGE formation, significantly improve the microstructure and biomechanical strength of diabetic bone and enhance bone fracture healing. Therefore, targeting AGEs may offer a strategy to regulate bone mineralization and microstructure, potentially preventing the onset of DBD.


Subject(s)
Bone Density , Diabetes Mellitus, Experimental , Glycation End Products, Advanced , Animals , Glycation End Products, Advanced/metabolism , Mice , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/pathology , Diabetes Mellitus, Experimental/complications , Biomineralization , Male , Mice, Inbred C57BL , Receptors, Leptin/metabolism , Receptors, Leptin/genetics , Bone and Bones/metabolism , Bone and Bones/pathology , Bone Diseases/pathology , Bone Diseases/metabolism , Disease Models, Animal , Collagen/metabolism , Diabetes Complications/metabolism , Diabetes Complications/pathology , Guanidines/pharmacology
20.
Int J Mol Sci ; 25(16)2024 Aug 11.
Article in English | MEDLINE | ID: mdl-39201444

ABSTRACT

Emerging evidence suggests that retinal neurodegeneration is an early event in the pathogenesis of diabetic retinopathy (DR), preceding the development of microvascular abnormalities. Here, we assessed the impact of neuroinflammation on the retina of diabetic-induced rats. For this aim we have used a two-photon microscope to image the photoreceptors (PRs) at different eccentricities in unstained retinas obtained from both control (N = 4) and pathological rats (N = 4). This technique provides high-resolution images where individual PRs can be identified. Within each image, every PR was located, and its transversal area was measured and used as an objective parameter of neuroinflammation. In control samples, the size of the PRs hardly changed with retinal eccentricity. On the opposite end, diabetic retinas presented larger PR transversal sections. The ratio of PRs suffering from neuroinflammation was not uniform across the retina. Moreover, the maximum anatomical resolving power (in cycles/deg) was also calculated. This presents a double-slope pattern (from the central retina towards the periphery) in both types of specimens, although the values for diabetic retinas were significantly lower across all retinal locations. The results show that chronic retinal inflammation due to diabetes leads to an increase in PR transversal size. These changes are not uniform and depend on the retinal location. Two-photon microscopy is a useful tool to accurately characterize and quantify PR inflammatory processes and retinal alterations.


Subject(s)
Diabetes Mellitus, Experimental , Diabetic Retinopathy , Animals , Diabetic Retinopathy/diagnostic imaging , Diabetic Retinopathy/pathology , Rats , Diabetes Mellitus, Experimental/pathology , Male , Photoreceptor Cells, Vertebrate/pathology , Disease Models, Animal , Retina/pathology , Retina/diagnostic imaging , Microscopy, Fluorescence, Multiphoton/methods , Microscopy/methods
SELECTION OF CITATIONS
SEARCH DETAIL