Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 17.818
Filter
1.
Biomaterials ; 313: 122804, 2025 Feb.
Article in English | MEDLINE | ID: mdl-39236631

ABSTRACT

Insulin resistance and pancreatic ß-cell dysfunction are the main pathogenesis of type 2 diabetes mellitus (T2DM). However, insulin therapy and diabetes medications do not effectively solve the two problems simultaneously. In this study, a biomimetic oral hydrogen nanogenerator that leverages the benefits of edible plant-derived exosomes and hydrogen therapy was constructed to overcome this dilemma by modulating gut microbiota and ameliorating oxidative stress and inflammatory responses. Hollow mesoporous silica (HMS) nanoparticles encapsulating ammonia borane (A) were used to overcome the inefficiency of H2 delivery in traditional hydrogen therapy, and exosomes originating from ginger (GE) were employed to enhance biocompatibility and regulate intestinal flora. Our study showed that HMS/A@GE not only considerably ameliorated insulin resistance and liver steatosis, but inhibited the dedifferentiation of islet ß-cell and enhanced pancreatic ß-cell proportion in T2DM model mice. In addition to its antioxidant and anti-inflammatory effects, HMS/A@GE augmented the abundance of Lactobacilli spp. and tryptophan metabolites, such as indole and indole acetic acid, which further activated the AhR/IL-22 pathway to improve intestinal-barrier function and metabolic impairments. This study offers a potentially viable strategy for addressing the current limitations of diabetes treatment by integrating gut-microbiota remodelling with antioxidant therapies.


Subject(s)
Antioxidants , Diabetes Mellitus, Type 2 , Gastrointestinal Microbiome , Insulin Resistance , Insulin-Secreting Cells , Nanoparticles , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/metabolism , Animals , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/drug therapy , Antioxidants/pharmacology , Gastrointestinal Microbiome/drug effects , Nanoparticles/chemistry , Mice , Male , Biomimetic Materials/chemistry , Biomimetic Materials/pharmacology , Mice, Inbred C57BL , Zingiber officinale/chemistry , Silicon Dioxide/chemistry , Exosomes/metabolism , Biomimetics/methods , Oxidative Stress/drug effects
2.
Commun Biol ; 7(1): 1267, 2024 Oct 05.
Article in English | MEDLINE | ID: mdl-39369076

ABSTRACT

Cellular bioenergetics and mitochondrial dynamics are crucial for the secretion of insulin by pancreatic beta cells in response to elevated levels of blood glucose. To elucidate the interactions between energy production and mitochondrial fission/fusion dynamics, we combine live-cell mitochondria imaging with biophysical-based modeling and graph-based network analysis. The aim is to determine the mechanism that regulates mitochondrial morphology and balances metabolic demands in pancreatic beta cells. A minimalistic differential equation-based model for beta cells is constructed that includes glycolysis, oxidative phosphorylation, calcium dynamics, and fission/fusion dynamics, with ATP synthase flux and proton leak flux as main regulators of mitochondrial dynamics. The model shows that mitochondrial fission occurs in response to hyperglycemia, starvation, ATP synthase inhibition, uncoupling, and diabetic conditions, in which the rate of proton leakage exceeds the rate of mitochondrial ATP synthesis. Under these metabolic challenges, the propensities of tip-to-tip fusion events simulated from the microscopy images of the mitochondrial networks are lower than those in the control group and prevent the formation of mitochondrial networks. The study provides a quantitative framework that couples bioenergetic regulation with mitochondrial dynamics, offering insights into how mitochondria adapt to metabolic challenges.


Subject(s)
Energy Metabolism , Insulin-Secreting Cells , Mitochondria , Mitochondrial Dynamics , Insulin-Secreting Cells/metabolism , Mitochondria/metabolism , Animals , Models, Biological , Mice , Adenosine Triphosphate/metabolism , Humans
3.
Recent Adv Drug Deliv Formul ; 18(4): 304-314, 2024.
Article in English | MEDLINE | ID: mdl-39356101

ABSTRACT

BACKGROUND: Autophagy plays a crucial role in modulating the proliferation of cancer diseases. However, the application of Naringenin (Nar), a compound with potential benefits against these diseases, has been limited due to its poor solubility and bioavailability. OBJECTIVE: This study aimed to develop solid lipid nanoparticles (Nar-SLNs) loaded with Nar to enhance their therapeutic impact. METHODS: In vitro experiments using Rin-5F cells exposed to Nar and Nar-SLNs were carried out to investigate the protective effects of Nar and its nanoformulation against the pancreatic cancer cell line of Rin-5F. RESULTS: Treatment with Nar and Nar-SLN led to an increase in autophagic markers (Akt, LC3, Beclin1, and ATG genes) and a decrease in the level of miR-21. Both Nar and Nar-SLN treatments inhibited cell proliferation and reduced the expression of autophagic markers. Notably, Nar-SLNs exhibited greater efficacy compared to free Nar. CONCLUSION: These findings suggest that SLNs effectively enhance the cytotoxic impact of Nar, making Nar-SLNs a promising candidate for suppressing or preventing Rin-5F cell growth.


Subject(s)
Autophagy , Cell Proliferation , Flavanones , Nanoparticles , Flavanones/pharmacology , Flavanones/administration & dosage , Flavanones/chemistry , Autophagy/drug effects , Nanoparticles/chemistry , Cell Proliferation/drug effects , Cell Line, Tumor , Animals , Rats , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/metabolism , Insulin-Secreting Cells/pathology , Lipids/chemistry , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/pathology , Pancreatic Neoplasms/metabolism , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/administration & dosage , Cell Survival/drug effects , Humans , Drug Carriers/chemistry , Liposomes
4.
Front Immunol ; 15: 1444639, 2024.
Article in English | MEDLINE | ID: mdl-39359722

ABSTRACT

Introduction: We reported that Ca2+-independent phospholipase A2ß (iPLA2ß)-derived lipids (iDLs) contribute to type 1 diabetes (T1D) onset. As CD4+ and CD8+ T cells are critical in promoting ß-cell death, we tested the hypothesis that iDL signaling from these cells participates in T1D development. Methods: CD4+ and CD8+ T cells from wild-type non-obese diabetic (NOD) and NOD.iPLA2ß+/- (NOD.HET) mice were administered in different combinations to immunodeficient NOD.scid. Results: In mice receiving only NOD T cells, T1D onset was rapid (5 weeks), incidence 100% by 20 weeks, and islets absent. In contrast, onset was delayed 1 week and incidence reduced 40%-50% in mice receiving combinations that included NOD.HET T cells. Consistently, islets from these non-diabetic mice were devoid of infiltrate and contained insulin-positive ß-cells. Reduced iPLA2ß led to decreased production of proinflammatory lipids from CD4+ T cells including prostaglandins and dihydroxyeicosatrienoic acids (DHETs), products of soluble epoxide hydrolase (sEH), and inhibition of their signaling decreased (by 82%) IFNγ+CD4+ cells abundance. However, only DHETs production was reduced from CD8+ T cells and was accompanied by decreases in sEH and granzyme B. Discussion: These findings suggest that differential select iDL signaling in CD4+ and CD8+ T cells contributes to T1D development, and that therapeutics targeting such signaling might be considered to counter T1D.


Subject(s)
CD4-Positive T-Lymphocytes , CD8-Positive T-Lymphocytes , Diabetes Mellitus, Type 1 , Mice, Inbred NOD , Signal Transduction , Animals , Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 1/metabolism , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Mice , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Insulin-Secreting Cells/metabolism , Insulin-Secreting Cells/immunology , Group VI Phospholipases A2/metabolism , Group VI Phospholipases A2/genetics , Lipid Metabolism , Mice, SCID , Female
5.
J Cell Mol Med ; 28(19): e70130, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39365284

ABSTRACT

Pancreatic ß-cell damage mediated by apoptosis is believed to be a main trigger of type 1 diabetes mellitus (T1DM), which is proposed as an organ-specific autoimmune disease mediated by T cells. Nonetheless, the fundamental origins of T1DM remain uncertain. Here, we illustrate that an increase in PLAGL1 expression induces ß-cell apoptosis, as evidenced by mitochondrial membrane impairment and nucleolar degradation. The gene expression levels from cDNA samples were determined using qRT-PCR method. Western blot and Co-immunoprecipitation were applied for protein expression and interactions, respectively. Flow cytometry and TUNEL assay were used to detect pancreatic ß cell apoptosis. Female NOD/LtJ mice with recent-onset T1DM has been used in in vivo studies. Glucose-stimulated insulin secretion (GSIS) and glucose tolerance test (GTT) method is used for islet function assessment. Haematoxylin and Eosin (H&E) and Immunohistochemistry (IHC) were performed to evalute histological improvement of islet beta. Subsequent cytoplasmic DNA accumulation triggers DNA senser, the cyclic guanosine monophosphate-AMP synthase (cGAS)-stimulator of interferon genes (STING) pathway. STING activation further stimulates downstream IRF3 and NF-kB pathways, thus boost type-I interferon signalling and NF-kB mediated inflammation. These findings elucidate a molecular mechanism linking PLAGL1 induced cell apoptosis to type-I interferon signalling and suggest a potential benefit for targeting cGAS/STING in T1DM treatment.


Subject(s)
Apoptosis , Insulin-Secreting Cells , Membrane Proteins , Nucleotidyltransferases , Animals , Nucleotidyltransferases/metabolism , Nucleotidyltransferases/genetics , Membrane Proteins/metabolism , Membrane Proteins/genetics , Mice , Insulin-Secreting Cells/metabolism , Insulin-Secreting Cells/pathology , Female , Signal Transduction , Cytoplasm/metabolism , DNA/metabolism , DNA/genetics , Humans , Transcription Factors/metabolism , Transcription Factors/genetics , Mice, Inbred NOD , Diabetes Mellitus, Type 1/metabolism , Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 1/pathology , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/genetics , Interferon Regulatory Factor-3/metabolism , Interferon Regulatory Factor-3/genetics , NF-kappa B/metabolism
7.
Signal Transduct Target Ther ; 9(1): 262, 2024 Oct 02.
Article in English | MEDLINE | ID: mdl-39353925

ABSTRACT

Type 2 diabetes (T2D) is a disease characterized by heterogeneously progressive loss of islet ß cell insulin secretion usually occurring after the presence of insulin resistance (IR) and it is one component of metabolic syndrome (MS), and we named it metabolic dysfunction syndrome (MDS). The pathogenesis of T2D is not fully understood, with IR and ß cell dysfunction playing central roles in its pathophysiology. Dyslipidemia, hyperglycemia, along with other metabolic disorders, results in IR and/or islet ß cell dysfunction via some shared pathways, such as inflammation, endoplasmic reticulum stress (ERS), oxidative stress, and ectopic lipid deposition. There is currently no cure for T2D, but it can be prevented or in remission by lifestyle intervention and/or some medication. If prevention fails, holistic and personalized management should be taken as soon as possible through timely detection and diagnosis, considering target organ protection, comorbidities, treatment goals, and other factors in reality. T2D is often accompanied by other components of MDS, such as preobesity/obesity, metabolic dysfunction associated steatotic liver disease, dyslipidemia, which usually occurs before it, and they are considered as the upstream diseases of T2D. It is more appropriate to call "diabetic complications" as "MDS-related target organ damage (TOD)", since their development involves not only hyperglycemia but also other metabolic disorders of MDS, promoting an up-to-date management philosophy. In this review, we aim to summarize the underlying mechanism, screening, diagnosis, prevention, and treatment of T2D, especially regarding the personalized selection of hypoglycemic agents and holistic management based on the concept of "MDS-related TOD".


Subject(s)
Diabetes Mellitus, Type 2 , Humans , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/therapy , Diabetes Mellitus, Type 2/pathology , Diabetes Mellitus, Type 2/metabolism , Insulin Resistance/genetics , Endoplasmic Reticulum Stress/genetics , Adult , Metabolic Syndrome/therapy , Metabolic Syndrome/genetics , Metabolic Syndrome/metabolism , Insulin-Secreting Cells/metabolism , Insulin-Secreting Cells/pathology , Dyslipidemias/genetics , Dyslipidemias/therapy , Dyslipidemias/pathology , Dyslipidemias/metabolism , Obesity/genetics , Obesity/therapy , Obesity/pathology , Oxidative Stress
8.
Nutr J ; 23(1): 119, 2024 Oct 01.
Article in English | MEDLINE | ID: mdl-39354480

ABSTRACT

BACKGROUND: Type 2 diabetes mellitus (T2DM) is a chronic condition characterized by insulin resistance and impaired insulin production, leading to elevated blood glucose levels. Curcumin, a polyphenolic compound from Curcuma longa, has shown potential in improving insulin sensitivity and reducing blood glucose levels, which may help mitigate type 2 diabetes progression. OBJECTIVE: To assess the efficacy of improving type 2 diabetes (T2DM). STUDY DESIGN: This randomized, double-blind, placebo-controlled trial included subjects (n = 272) with criteria for type 2 diabetes. METHODS: All subjects were randomly assigned to receive curcumin (1500 mg/day) or placebo with blind labels for 12 months. To assess the improvement of T2DM after curcumin treatments body weight and body mass index, fasting plasma glucose, glycosylated hemoglobin A1c, ß-cell function (homeostasis model assessment [HOMA-ß]), insulin resistance (HOMA-IR), insulin, adiponectin, and leptin were monitored at the baseline and at 3-, 6-, 9-, and 12-month visits during the course of intervention. RESULTS: After 12 months of treatment, the curcumin-treated group showed a significant decrease in fasting blood glucose (115.49 vs.130.71; P < 0.05), HbA1c (6.12 vs. 6.47; P < 0.05). In addition, the curcumin-treated group showed a better overall function of ß-cells, with higher HOMA-ß (136.20 vs. 105.19; P < 0.01) The curcumin-treated group showed a lower level of HOMA-IR (4.86 vs. 6.04; P < 0.001) and higher adiponectin (14.51 vs. 10.36; P < 0.001) when compared to the placebo group. The curcumin-treated group also showed a lower level of leptin (9.42 vs. 20.66; P < 0.001). Additionally, body mass index was lowered (25.9 4 vs.29.34), with a P value of 0.001. CONCLUSIONS: A 12-month curcumin intervention in type 2 diabetes patients shows a significant glucose-lowering effect. Curcumin treatment appeared to improve the overall function of ß-cells and reduce both insulin resistance and body weight, with very minor adverse effects. Curcumin intervention in obese patients with type 2 diabetes may be beneficial. TRIAL REGISTRATION: Thai clinical trials regentrify no.20140303003.


Subject(s)
Blood Glucose , Body Mass Index , Curcumin , Diabetes Mellitus, Type 2 , Glycated Hemoglobin , Insulin Resistance , Insulin-Secreting Cells , Insulin , Obesity , Humans , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/complications , Curcumin/pharmacology , Curcumin/administration & dosage , Male , Female , Double-Blind Method , Middle Aged , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/metabolism , Blood Glucose/drug effects , Blood Glucose/metabolism , Obesity/drug therapy , Obesity/complications , Insulin/blood , Adiponectin/blood , Plant Extracts/pharmacology , Plant Extracts/administration & dosage , Leptin/blood , Adult , Curcuma , Body Weight/drug effects
9.
Adv Anat Embryol Cell Biol ; 239: 57-90, 2024.
Article in English | MEDLINE | ID: mdl-39283482

ABSTRACT

The existence of functionally diverse and plastic ß cells in islets of Langerhans has been reported since the 1980s. Recently, high-resolution technologies have advanced our understanding of ß-cell heterogeneity and plasticity. Here, we define plasticity broadly as dynamic changes in cellular phenotypes and heterogeneity as differences in cellular behaviors. Individual ß cells react differently to environmental challenges and act together to maintain ß-cell mass and glucose homeostasis within a narrow range of 70-140 mg/dL. During the progress of diabetes, this elaborate balance is disrupted, and a lack of ß-cell compensation leads to dysregulated blood glucose. In this chapter, we assess ß-cell stress that instigates increased ß-cell heterogeneity and adaptive ß-cell responses such as proliferation, dedifferentiation, maturity, and insulin secretion. We also discuss the maturity, electrical activity, and insulin secretion of well-characterized ß-cell subgroups. Finally, we touch upon the plasticity of other non-ß pancreatic cells and their cooperation with ß cells to maintain homeostasis.


Subject(s)
Cell Plasticity , Insulin-Secreting Cells , Insulin-Secreting Cells/physiology , Insulin-Secreting Cells/metabolism , Humans , Animals , Insulin Secretion , Insulin/metabolism , Homeostasis
10.
Adv Anat Embryol Cell Biol ; 239: 157-197, 2024.
Article in English | MEDLINE | ID: mdl-39283486

ABSTRACT

Maternal nutrition and metabolic health status during pregnancy are critical factors that shape the life-long health trajectory of offspring. Altered nutrition during specific times of development in utero can lead to functional changes in tissues such as the pancreatic ß-cells, predisposing those tissues to metabolic diseases and Type 2 diabetes that manifest later in life. This chapter will focus on the role of pregnancy complications with altered nutrition during gestation in the maladaptive programming of ß-cell mass and function in the offspring.


Subject(s)
Insulin-Secreting Cells , Female , Pregnancy , Insulin-Secreting Cells/metabolism , Humans , Maternal Nutritional Physiological Phenomena , Animals , Prenatal Exposure Delayed Effects/metabolism , Nutritional Status , Pregnancy Complications , Diabetes Mellitus, Type 2/metabolism
11.
Adv Anat Embryol Cell Biol ; 239: 91-115, 2024.
Article in English | MEDLINE | ID: mdl-39283483

ABSTRACT

The pancreas has been considered a non-regenerative organ. ß cells lost in diabetes are not replaced due to the inability of the pancreas to regenerate. However, ample evidence generated in the last few decades using murine models has demonstrated that the pancreas has a remarkable plasticity wherein differentiated cells can change cell fate toward a ß-like cell phenotype. Although this process is observed after using rather artificial stimuli and the conversion efficiency is very limited, these findings have shed some light on novel pathways for ß-cell regeneration. In this chapter, we will summarize the different cellular interconversion processes described to date, the experimental details and molecular regulation of such interconversions, and the genomic technologies that have allowed the identification of potential new ways to generate ß cells.


Subject(s)
Cell Plasticity , Insulin-Secreting Cells , Regeneration , Animals , Insulin-Secreting Cells/physiology , Insulin-Secreting Cells/cytology , Regeneration/physiology , Humans , Cell Plasticity/physiology , Pancreas/physiology , Pancreas/cytology , Cell Differentiation , Mice
12.
Adv Anat Embryol Cell Biol ; 239: 117-139, 2024.
Article in English | MEDLINE | ID: mdl-39283484

ABSTRACT

The pancreatic ß cells are at the hub of myriad signals to regulate the secretion of an adequate amount of insulin needed to re-establish postprandial euglycemia. The ß cell possesses sophisticated metabolic enzymes and a variety of extracellular receptors and channels that amplify insulin secretion in response to autocrine, paracrine, and neurohormonal signals. Considerable research has been undertaken to decipher the mechanisms regulating insulin secretion. While the triggering pathway induced by glucose is needed to initiate the exocytosis process, multiple other stimuli modulate the insulin secretion response. This chapter will discuss the recent advances in understanding the role of the diverse glucose- and fatty acid-metabolic coupling factors in amplifying insulin secretion. It will also highlight the intracellular events linking the extracellular receptors and channels to insulin secretion amplification. Understanding these mechanisms provides new insights into learning more about the etiology of ß-cell failure and paves the way for developing new therapeutic strategies for type 2 diabetes.


Subject(s)
Insulin Secretion , Insulin-Secreting Cells , Insulin , Humans , Insulin-Secreting Cells/metabolism , Insulin/metabolism , Animals , Glucose/metabolism , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/genetics , Signal Transduction , Fatty Acids/metabolism
13.
Adv Anat Embryol Cell Biol ; 239: 141-155, 2024.
Article in English | MEDLINE | ID: mdl-39283485

ABSTRACT

Pancreatic δ cells act locally to repress both insulin and glucagon secretion. Because they are a rare cell type, experimentation examining δ-cell function and control has lagged that of the more abundant α and ß cells. Emerging evidence, enabled partly by developing single-cell technology, demonstrates that δ-cell function is, in part, directed by δ cells but that δ cells also have intrinsic control. The contribution of these cells to overall glucose homeostasis and diabetes onset and progression is still unclear. However, they regulate both α and ß cells, both of which are dysfunctional in diabetes, and their numbers are disrupted in humans with diabetes and in multiple animal models of diabetes, suggesting δ cells are a pivotal character in both health and disease.


Subject(s)
Insulin-Secreting Cells , Humans , Animals , Insulin-Secreting Cells/physiology , Diabetes Mellitus , Somatostatin-Secreting Cells/metabolism , Insulin/metabolism , Glucagon-Secreting Cells/metabolism , Glucagon/metabolism
14.
Nutrients ; 16(17)2024 Aug 27.
Article in English | MEDLINE | ID: mdl-39275180

ABSTRACT

Dysfunction or loss of pancreatic ß cells can cause insulin deficiency and impaired glucose regulation, resulting in conditions like type 2 diabetes. The ATP-binding cassette transporter A1 (ABCA1) plays a key role in the reverse cholesterol transport system, and its decreased expression is associated with pancreatic ß cell lipotoxicity, resulting in abnormal insulin synthesis and secretion. Increased glutamate release can cause glucotoxicity in ß cells, though the detailed mechanisms remain unclear. This study investigated the effect of N-methyl-D-aspartic acid (NMDA) on ABCA1 expression in INS-1 cells and primary pancreatic islets to elucidate the signaling mechanisms that suppress insulin secretion. Using Western blotting, microscopy, and biochemical analyses, we found that NMDA activated the mitogen-activated protein kinase (MEK)-dependent pathway, suppressing ABCA1 protein and mRNA expression. The MEK-specific inhibitor PD98059 restored ABCA1 promoter activity, indicating the involvement of the extracellular signal-regulated kinase (MEK/ERK) pathway. Furthermore, we identified the liver X receptor (LXR) as an effector transcription factor in NMDA regulation of ABCA1 transcription. NMDA treatment increased cholesterol and triglyceride levels while decreasing insulin secretion, even under high-glucose conditions. These effects were abrogated by treatment with PD98059. This study reveals that NMDA suppresses ABCA1 expression via the MEK/ERK/LXR pathway, providing new insights into the pathological suppression of insulin secretion in pancreatic ß cells and emphasizing the importance of investigating the role of NMDA in ß cell dysfunction.


Subject(s)
ATP Binding Cassette Transporter 1 , Insulin-Secreting Cells , Liver X Receptors , MAP Kinase Signaling System , N-Methylaspartate , Insulin-Secreting Cells/metabolism , Insulin-Secreting Cells/drug effects , Animals , ATP Binding Cassette Transporter 1/metabolism , ATP Binding Cassette Transporter 1/genetics , N-Methylaspartate/pharmacology , Rats , Liver X Receptors/metabolism , MAP Kinase Signaling System/drug effects , Cholesterol/metabolism , Insulin/metabolism , Insulin Secretion/drug effects , Male , Extracellular Signal-Regulated MAP Kinases/metabolism , Cell Line
16.
Front Endocrinol (Lausanne) ; 15: 1422279, 2024.
Article in English | MEDLINE | ID: mdl-39239092

ABSTRACT

Type 1 Diabetes (T1D) is a chronic metabolic disease resulting from insulin deficiency due to autoimmune loss of pancreatic ß cells. In addition to ß cell destruction, it is now accepted that ß cell stress and dysfunction, such as senescence, plays a crucial role in the development of the disease. Accumulation of senescent ß cells occurs during development of T1D in humans and contributes to the progression of T1D in the nonobese diabetic (NOD) mouse model. Senescent ß cells are thought to exacerbate the inflammatory response within the islets by production and secretion of senescence-associated secretory phenotype (SASP). Extracellular vesicles (EVs) from ß cells have been shown to carry protein and microRNAs (miRNAs), influencing cellular signaling and may contribute to the development of T1D but it remains to be addressed how senescence impacts ß cell EV cargo. In this minireview, we discuss emerging evidence that EV cargo proteins and miRNAs associated with senescence could contribute to the development of T1D and could suggest potential biomarkers and therapeutic targets for the regulation of SASP and elimination of senescent ß cells in T1D. Future investigation exploring the intricate relationship between ß cell senescence, EVs and miRNAs could pave the way for the development of novel diagnostic techniques and therapeutic interventions.


Subject(s)
Cellular Senescence , Diabetes Mellitus, Type 1 , Extracellular Vesicles , Insulin-Secreting Cells , Diabetes Mellitus, Type 1/metabolism , Diabetes Mellitus, Type 1/pathology , Humans , Extracellular Vesicles/metabolism , Insulin-Secreting Cells/metabolism , Insulin-Secreting Cells/pathology , Animals , MicroRNAs/metabolism , MicroRNAs/genetics , Senescence-Associated Secretory Phenotype
17.
Int J Med Sci ; 21(11): 2081-2093, 2024.
Article in English | MEDLINE | ID: mdl-39239539

ABSTRACT

Insulin resistance is the primary contributor to the disruption in glucose homeostasis in the body, playing a significant causative role in many metabolic diseases. Insulin resistance is characterized by compensatory insulin secretion and reduced insulin responsiveness in target organs. Dysregulation of the interaction between insulin-secreting cells and insulin-responsive target organs is an important factor driving the progression of insulin resistance. Circulating endocrine hormones are important mediators mediating the interaction between insulin-secreting cells and insulin-responsive target organs. In addition to the classical hormones secreted by endocrine glands and organ-specific hormones secreted by metabolism-related organs (adipose tissue, muscle, liver, etc.), extracellular vesicles have been recognized as a novel class of endocrine hormones with a complex composition. Extracellular vesicles can transport signaling molecules, such as miRNAs and LncRNAs, to vital organs related to insulin resistance, in a manner akin to conventional hormones. The significant role in regulating the development of insulin resistance underscores the increasing interest in extracellular vesicles as essential contributors to this process. In this review, we summarize the three types of hormones (classical hormones, organokines and extracellular vesicles) that play a regulatory role in insulin resistance, and focus on the novel endocrine hormones, extracellular vesicles, to elaborate the mechanism of extracellular vesicles' regulation of insulin resistance progress from two aspects: the impact on insulin-secreting cells and the influence on insulin-responsive target organs. In addition, this paper outlines the clinical applications of extracellular vesicles in insulin resistance. A comprehensive understanding of the regulatory mechanisms and diagnostic status of the inter-organ network in insulin resistance has great potential to advance targeted therapeutic interventions and diagnostic markers, thereby benefiting both the prevention and treatment of insulin resistance.


Subject(s)
Extracellular Vesicles , Insulin Resistance , Humans , Extracellular Vesicles/metabolism , Insulin-Secreting Cells/metabolism , Insulin-Secreting Cells/physiology , Insulin/metabolism , Hormones/metabolism , Animals , Adipose Tissue/metabolism , MicroRNAs/metabolism , MicroRNAs/genetics
18.
Sci Rep ; 14(1): 20653, 2024 09 04.
Article in English | MEDLINE | ID: mdl-39232042

ABSTRACT

Stem cell-derived ß-cells (SC-BCs) represent a potential source for curing diabetes. To date, in vitro generated SC-BCs display an immature phenotype and lack important features in comparison to their bona-fide counterparts. Transplantation into a living animal promotes SC-BCs maturation, indicating that components of the in vivo microenvironment trigger final SC-BCs development. Here, we investigated whether cues of the pancreas specific extracellular matrix (ECM) can improve the differentiation of human induced pluripotent stem cells (hiPSCs) towards ß-cells in vitro. To this aim, a pancreas specific ECM (PanMa) hydrogel was generated from decellularized porcine pancreas and its effect on the differentiation of hiPSC-derived pancreatic hormone expressing cells (HECs) was tested. The hydrogel solidified upon neutralization at 37 °C with gelation kinetics similar to Matrigel. Cytocompatibility of the PanMa hydrogel was demonstrated for a culture duration of 21 days. Encapsulation and culture of HECs in the PanMa hydrogel over 7 days resulted in a stable gene and protein expression of most ß-cell markers, but did not improve ß-cell identity. In conclusion, the study describes the production of a PanMa hydrogel, which provides the basis for the development of ECM hydrogels that are more adapted to the demands of SC-BCs.


Subject(s)
Cell Differentiation , Extracellular Matrix , Hydrogels , Induced Pluripotent Stem Cells , Insulin-Secreting Cells , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Humans , Hydrogels/chemistry , Insulin-Secreting Cells/cytology , Insulin-Secreting Cells/metabolism , Extracellular Matrix/metabolism , Animals , Swine , Pancreas/cytology , Pancreas/metabolism , Cell Culture Techniques/methods , Cells, Cultured
19.
Int J Nanomedicine ; 19: 9575-9596, 2024.
Article in English | MEDLINE | ID: mdl-39296939

ABSTRACT

Background: The depletion of ß cell mass is widely recognized as a significant contributor to the progression of type 2 diabetes mellitus (T2DM). Exosomes derived from mesenchymal stem cells (MSC-EXOs) hold promise as cell-free therapies for treating T2DM. However, the precise effects and mechanisms through which MSC-EXO affects ß cell function remain incompletely understood, and the limited ability of MSC-EXO to target ß cells and the short blood circulation time hampers its therapeutic effectiveness. Methods: The effects of MSC-EXO were investigated in T2DM mice induced by a high-fat diet combined with STZ. Additionally, the high glucose-stimulated INS-1 cell line was used to investigate the potential mechanism of MSC-EXO. Michael addition reaction-mediated chemical coupling was used to modify the surface of the exosome membrane with a ß-cell-targeting aptamer and polyethylene glycol (PEG). The ß-cell targeting and blood circulation time were evaluated, and whether this modification enhanced the islet-protective effect of MSC-EXO was further analyzed. Results: We observed that the therapeutic effects of MSC-EXO on T2DM manifested through the reduction of random blood glucose levels, enhancement of glucose and insulin tolerance, and increased insulin secretion. These effects were achieved by augmenting ß cell mass via inhibiting nuclear factor erythroid 2-related factor 2 (NRF2)-mediated ferroptosis. Mechanistically, MSC-EXOs play a role in the NRF2-mediated anti-ferroptosis mechanism by transporting active proteins that are abundant in the AKT and ERK pathways. Moreover, compared to MSC-EXOs, aptamer- and PEG-modified exosomes (Apt-EXOs) were more effective in islet protection through PEG-mediated cycle prolongation and aptamer-mediated ß-cell targeting. Conclusion: MSC-EXO suppresses NRF2-mediated ferroptosis by delivering bioactive proteins to regulate the AKT/ERK signaling pathway, thereby improving the function and quantity of ß cells. Additionally, Apt-EXO may serve as a novel drug carrier for islet-targeted therapy.


Subject(s)
Exosomes , Ferroptosis , Insulin-Secreting Cells , Mesenchymal Stem Cells , NF-E2-Related Factor 2 , Polyethylene Glycols , Animals , Exosomes/metabolism , Exosomes/chemistry , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism , NF-E2-Related Factor 2/metabolism , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/metabolism , Mice , Ferroptosis/drug effects , Polyethylene Glycols/chemistry , Polyethylene Glycols/pharmacology , Male , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/therapy , Diabetes Mellitus, Experimental/therapy , Mice, Inbred C57BL , Cell Line , Aptamers, Nucleotide/chemistry , Aptamers, Nucleotide/pharmacology , Diet, High-Fat , Blood Glucose/metabolism
20.
Islets ; 16(1): 2406041, 2024 Dec 31.
Article in English | MEDLINE | ID: mdl-39298538

ABSTRACT

BACKGROUND: Islet or ß-cell transplantation is a therapeutical approach to substitute the insulin-producing cells which are abolished in type 1 diabetes mellitus. The shortage of human islets as well as the complicated and costly isolation process limit the application of these techniques in daily clinical practice. EndoC-ßH is a human ß-cell line that readily forms aggregates termed pseudoislets, providing an alternative to primary human islets or ß-cells. METHODS: EndoC-ßH3 cells were seeded and incubated to form pseudoislets. Their insulin secretion was analyzed by ELISA and compared with cell monolayers. Pseudoislets were transplanted into streptozotocin-treated NMRi nu/nu mice. Blood glucose was monitored before and after transplantation and compared with wild types. Grafts were analyzed by immunohistology. RESULTS: This study shows that EndoC-ßH cells are able to form pseudoislets by aggregation, leading to an enhanced glucose stimulated insulin secretion in vitro. These pseudoislets were then successfully transplanted into the livers of diabetic mice and produced insulin in vitro. Blood glucose levels of the streptozocin-treated recipient mice were significantly decreased when compared to pre-transplantation and matched the levels found in control mice. CONCLUSION: We suggest pseudoislets aggregated from EndoC-ßH cells as a valuable and promising model for islet transplantation research.


Subject(s)
Blood Glucose , Diabetes Mellitus, Experimental , Insulin-Secreting Cells , Insulin , Islets of Langerhans Transplantation , Animals , Diabetes Mellitus, Experimental/therapy , Mice , Islets of Langerhans Transplantation/methods , Humans , Insulin-Secreting Cells/metabolism , Insulin/metabolism , Blood Glucose/metabolism , Insulin Secretion , Cell Line , Mice, Nude , Male , Cell Aggregation , Diabetes Mellitus, Type 1/therapy , Diabetes Mellitus, Type 1/surgery , Diabetes Mellitus, Type 1/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL