Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 17.250
Filter
1.
Mol Biol Rep ; 51(1): 711, 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38824245

ABSTRACT

BACKGROUND: Diabetes is a chronic metabolic disease that affects many parts of the body. Considering diabetes as a beta cells' defect and loss, the focus is on finding mechanisms and compounds involved in stimulating the function and regeneration of pancreatic ß-cells. DNA methylation as an epigenetic mechanism plays a pivotal role in the ß-cells' function and development. Considering the regenerative and anti-diabetic effects of Rosa canina extract, this study aimed to assess the methylation levels of Pdx-1, Pax-4, and Ins-1 genes in diabetic rats treated with Rosa Canina extract. METHODS AND RESULTS: Streptozotocin-induced diabetic rats were used to evaluate the frequency of Pdx-1, Pax-4, and Ins-1 gene methylation. Treatment groups were exposed to Rosa canina as spray-dried and decoction extracts. Following blood glucose measurement, pancreatic DNA was extracted and bisulfited. Genes' methylation was measured using MSP-PCR and qRT-PCR techniques. Oral administration of Rosa canina extracts significantly reduced blood sugar levels in diabetic rats compared to the control group. The methylation levels of the Pdx-1, Pax-4, and Ins-1 genes promoter in streptozotocin-induced diabetic rats increased compared to the control rats while, the treatment of diabetic rats with Rosa canina extracts, spray-dried samples especially, led to a decreased methylation in these genes. CONCLUSION: The results of this study showed that Rosa canina extract as a spray-dried sample could be effective in treating diabetes by regulating the methylation of genes including Pdx-1, Pax-4, and Ins-1 involved in the activity and regeneration of pancreatic islet cells.


Subject(s)
Blood Glucose , DNA Methylation , Diabetes Mellitus, Experimental , Plant Extracts , Rosa , Trans-Activators , Animals , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Experimental/drug therapy , Rosa/chemistry , DNA Methylation/drug effects , DNA Methylation/genetics , Rats , Plant Extracts/pharmacology , Male , Trans-Activators/genetics , Trans-Activators/metabolism , Blood Glucose/metabolism , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Pancreas/drug effects , Pancreas/metabolism , Pancreas/pathology , Streptozocin , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/metabolism , Promoter Regions, Genetic/drug effects , Promoter Regions, Genetic/genetics , Paired Box Transcription Factors/genetics , Paired Box Transcription Factors/metabolism , Insulin/metabolism
2.
Drug Des Devel Ther ; 18: 1785-1797, 2024.
Article in English | MEDLINE | ID: mdl-38828020

ABSTRACT

Objective: Pancreatic surgeries inherently cause ischemia-reperfusion (IR) injury, affecting not only the pancreas but also distant organs. This study was conducted to explore the potential use of dexmedetomidine, a sedative with antiapoptotic, anti-inflammatory, and antioxidant properties, in mitigating the impacts of pancreatic IR on kidney and liver tissues. Methods: A total of 24 rats were randomly divided into four groups: control (C), dexmedetomidine (D), ischemia reperfusion (IR), and dexmedetomidine ischemia reperfusion (D-IR). Pancreatic ischemia was induced in the IR and D-IR groups. Dexmedetomidine was administered intraperitoneally to the D and D-IR groups. Liver and kidney tissue samples were subjected to microscopic examinations after hematoxylin and eosin staining. The levels of thiobarbituric acid reactive substances (TBARS), aryllesterase (AES), catalase (CAT), and glutathione S-transferase (GST) enzyme activity were assessed in liver and kidney tissues. The serum levels of aspartate aminotransferase (AST), alanine aminotransferase (ALT), blood urea nitrogen (BUN), and creatinine were measured. Results: A comparison of the groups revealed that the IR group exhibited significantly elevated TBARS (p < 0.0001), AES (p = 0.004), and CAT enzyme activity (p < 0.0001) levels in the liver and kidney compared to groups C and D. Group D-IR demonstrated notably reduced histopathological damage (p < 0.05) and low TBARS (p < 0.0001), AES (p = 0.004), and CAT enzyme activity (p < 0.0001) in the liver and kidney as well as low AST and ALT activity levels (p < 0.0001) in the serum compared to the IR group. Conclusion: The preemptive administration of dexmedetomidine before pancreatic IR provides significant protection to kidney and liver tissues, as evidenced by the histopathological and biochemical parameters in this study. The findings underscored the potential therapeutic role of dexmedetomidine in mitigating the multiorgan damage associated with pancreatic surgeries.


Subject(s)
Dexmedetomidine , Kidney , Liver , Pancreas , Reperfusion Injury , Animals , Reperfusion Injury/drug therapy , Reperfusion Injury/pathology , Reperfusion Injury/metabolism , Dexmedetomidine/pharmacology , Dexmedetomidine/administration & dosage , Rats , Kidney/drug effects , Kidney/pathology , Kidney/metabolism , Liver/drug effects , Liver/pathology , Liver/metabolism , Male , Pancreas/drug effects , Pancreas/pathology , Pancreas/metabolism , Rats, Sprague-Dawley
3.
BMC Genomics ; 25(1): 553, 2024 Jun 03.
Article in English | MEDLINE | ID: mdl-38831310

ABSTRACT

Development of the human pancreas requires the precise temporal control of gene expression via epigenetic mechanisms and the binding of key transcription factors. We quantified genome-wide patterns of DNA methylation in human fetal pancreatic samples from donors aged 6 to 21 post-conception weeks. We found dramatic changes in DNA methylation across pancreas development, with > 21% of sites characterized as developmental differentially methylated positions (dDMPs) including many annotated to genes associated with monogenic diabetes. An analysis of DNA methylation in postnatal pancreas tissue showed that the dramatic temporal changes in DNA methylation occurring in the developing pancreas are largely limited to the prenatal period. Significant differences in DNA methylation were observed between males and females at a number of autosomal sites, with a small proportion of sites showing sex-specific DNA methylation trajectories across pancreas development. Pancreas dDMPs were not distributed equally across the genome and were depleted in regulatory domains characterized by open chromatin and the binding of known pancreatic development transcription factors. Finally, we compared our pancreas dDMPs to previous findings from the human brain, identifying evidence for tissue-specific developmental changes in DNA methylation. This study represents the first systematic exploration of DNA methylation patterns during human fetal pancreas development and confirms the prenatal period as a time of major epigenomic plasticity.


Subject(s)
DNA Methylation , Pancreas , Humans , Pancreas/metabolism , Pancreas/embryology , Female , Male , Gene Expression Regulation, Developmental , CpG Islands , Epigenesis, Genetic , Genome, Human , Fetus/metabolism
4.
J Oleo Sci ; 73(5): 717-727, 2024.
Article in English | MEDLINE | ID: mdl-38692894

ABSTRACT

The anti-diabetic effect of Ficus carica (Fig) seed oil was investigated. 4 groups with 6 rats in each group were used in the experiment as control, diabetes (45 mg/kg streptozotocin), fig seed oil (FSO) (6 mL/ kg/day/rat by gavage) and diabetes+FSO groups. Glucose, urea, creatinine, ALT, AST, GSH, AOPP and MDA analyses were done. Pancreatic tissues were examined histopathologically. When fig seed oil was given to the diabetic group, the blood glucose level decreased. In the diabetes+FSO group, serum urea, creatinine, AOPP, MDA levels and ALT and AST activities decreased statistically significantly compared to the diabetes group, while GSH levels increased significantly, histopathological, immunohistochemical, and immunofluorescent improvements were observed. It has been shown for the first time that FSO has positive effects on blood glucose level and pancreatic health. It can be said that the protective effect of fig seed oil on tissues may be due to its antioxidant activity.


Subject(s)
Antioxidants , Blood Glucose , Diabetes Mellitus, Experimental , Ficus , Hypoglycemic Agents , Pancreas , Plant Oils , Seeds , Streptozocin , Animals , Ficus/chemistry , Diabetes Mellitus, Experimental/drug therapy , Plant Oils/pharmacology , Plant Oils/isolation & purification , Seeds/chemistry , Hypoglycemic Agents/pharmacology , Hypoglycemic Agents/isolation & purification , Blood Glucose/metabolism , Male , Pancreas/drug effects , Pancreas/pathology , Pancreas/metabolism , Antioxidants/pharmacology , Rats , Rats, Wistar , Creatinine/blood
5.
Front Endocrinol (Lausanne) ; 15: 1332895, 2024.
Article in English | MEDLINE | ID: mdl-38694937

ABSTRACT

Background: More than 700 million people worldwide suffer from diseases of the pancreas, such as diabetes, pancreatitis and pancreatic cancer. Often dysregulation of potassium (K+) channels, co-transporters and pumps can promote development and progression of many types of these diseases. The role of K+ transport system in pancreatic cell homeostasis and disease development remains largely unexplored. Potassium isotope analysis (δ41K), however, might have the potential to detect minute changes in metabolic processes relevant for pancreatic diseases. Methods: We assessed urinary K isotope composition in a case-control study by measuring K concentrations and δ41K in spot urines collected from patients diagnosed with pancreatic cancer (n=18), other pancreas-related diseases (n=14) and compared those data to healthy controls (n=16). Results: Our results show that urinary K+ levels for patients with diseased pancreas (benign and pancreatic cancer) are significantly lower than the healthy controls. For δ41K, the values tend to be higher for individuals with pancreatic cancer (mean δ41K = -0.58 ± 0.33‰) than for healthy individuals (mean δ41K = -0.78 ± 0.19‰) but the difference is not significant (p=0.08). For diabetics, urinary K+ levels are significantly lower (p=0.03) and δ41K is significantly higher (p=0.009) than for the healthy controls. These results suggest that urinary K+ levels and K isotopes can help identify K disturbances related to diabetes, an associated factors of all-cause mortality for diabetics. Conclusion: Although the K isotope results should be considered exploratory and hypothesis-generating and future studies should focus on larger sample size and δ41K analysis of other K-disrupting diseases (e.g., chronic kidney disease), our data hold great promise for K isotopes as disease marker.


Subject(s)
Diabetes Mellitus , Pancreatic Neoplasms , Potassium , Humans , Pancreatic Neoplasms/urine , Male , Female , Case-Control Studies , Middle Aged , Aged , Potassium/urine , Diabetes Mellitus/urine , Diabetes Mellitus/metabolism , Adult , Pancreas/metabolism , Isotopes/urine
6.
BMC Gastroenterol ; 24(1): 151, 2024 May 02.
Article in English | MEDLINE | ID: mdl-38698325

ABSTRACT

BACKGROUND: Acute pancreatitis (AP) is a prevalent exocrine inflammatory disorder of the pancreas characterized by pancreatic inflammation and injury to acinar cells. Vitamin B6 (VB6) is a vital nutrient that plays a significant role in preserving human health and has anti-inflammatory and anti-apoptotic effects. METHODS: This study aimed to explore the potential pancreatic protective effects of VB6 in mitigating pancreatic inflammation and apoptosis induced by taurocholate sodium (TLCS) in an AP model and to assess the underlying mechanism of action. AP was induced in Sprague‒Dawley (SD) rats through TLCS administration and lipopolysaccharide (LPS)-treated AR42J cells, followed by treatment with VB6. RESULTS: Various parameters associated with AP were assessed in both plasma and pancreatic tissues. VB6 has been shown to ameliorate the severity of AP through various mechanisms. It effectively reduces the levels of serum amylase, lipase, and inflammatory factors, thereby mitigating histological injury to the pancreas. Moreover, VB6 inhibited pancreatic apoptosis by downregulating bax expression and up-regulating Bcl2 expression in TLCS-treated rats. Additionally, VB6 suppressed the expression of caspase3. The anti-inflammatory and anti-apoptotic effects of VB6 observed in LPS-treated AR42J cells are consistent with those observed in a rat model of AP. CONCLUSIONS: These results suggest that VB6 exerts anti-inflammatory and anti-apoptotic effects through inhibition of the caspase3 signaling pathway and has a protective effect against AP.


Subject(s)
Apoptosis , Caspase 3 , Lipopolysaccharides , Pancreatitis , Rats, Sprague-Dawley , Signal Transduction , Taurocholic Acid , Vitamin B 6 , Animals , Pancreatitis/drug therapy , Pancreatitis/metabolism , Pancreatitis/pathology , Pancreatitis/chemically induced , Signal Transduction/drug effects , Apoptosis/drug effects , Caspase 3/metabolism , Rats , Vitamin B 6/pharmacology , Vitamin B 6/therapeutic use , Male , Amylases/blood , Pancreas/pathology , Pancreas/drug effects , Pancreas/metabolism , Disease Models, Animal , Anti-Inflammatory Agents/pharmacology , Acute Disease , bcl-2-Associated X Protein/metabolism , Lipase/metabolism , Lipase/blood , Proto-Oncogene Proteins c-bcl-2/metabolism
7.
Development ; 151(9)2024 May 01.
Article in English | MEDLINE | ID: mdl-38727565

ABSTRACT

Proper embryonic development depends on the timely progression of a genetic program. One of the key mechanisms for achieving precise control of developmental timing is to use gene expression oscillations. In this Review, we examine how gene expression oscillations encode temporal information during vertebrate embryonic development by discussing the gene expression oscillations occurring during somitogenesis, neurogenesis, myogenesis and pancreas development. These oscillations play important but varied physiological functions in different contexts. Oscillations control the period of somite formation during somitogenesis, whereas they regulate the proliferation-to-differentiation switch of stem cells and progenitor cells during neurogenesis, myogenesis and pancreas development. We describe the similarities and differences of the expression pattern in space (i.e. whether oscillations are synchronous or asynchronous across neighboring cells) and in time (i.e. different time scales) of mammalian Hes/zebrafish Her genes and their targets in different tissues. We further summarize experimental evidence for the functional role of their oscillations. Finally, we discuss the outstanding questions for future research.


Subject(s)
Embryonic Development , Gene Expression Regulation, Developmental , Somites , Animals , Embryonic Development/genetics , Humans , Somites/metabolism , Somites/embryology , Muscle Development/genetics , Neurogenesis/genetics , Neurogenesis/physiology , Pancreas/embryology , Pancreas/metabolism , Cell Differentiation/genetics
8.
Cell Death Dis ; 15(5): 348, 2024 May 20.
Article in English | MEDLINE | ID: mdl-38769308

ABSTRACT

Regenerating gene family member 4 (Reg4) has been implicated in acute pancreatitis, but its precise functions and involved mechanisms have remained unclear. Herein, we sought to investigate the contribution of Reg4 to the pathogenesis of pancreatitis and evaluate its therapeutic effects in experimental pancreatitis. In acute pancreatitis, Reg4 deletion increases inflammatory infiltrates and mitochondrial cell death and decreases autophagy recovery, which are rescued by the administration of recombinant Reg4 (rReg4) protein. In chronic pancreatitis, Reg4 deficiency aggravates inflammation and fibrosis and inhibits compensatory cell proliferation. Moreover, C-X-C motif ligand 12 (CXCL12)/C-X-C motif receptor 4 (CXCR4) axis is sustained and activated in Reg4-deficient pancreas. The detrimental effects of Reg4 deletion are attenuated by the administration of the approved CXCR4 antagonist plerixafor (AMD3100). Mechanistically, Reg4 mediates its function in pancreatitis potentially via binding its receptor exostosin-like glycosyltransferase 3 (Extl3). In conclusion, our findings suggest that Reg4 exerts a therapeutic effect during pancreatitis by limiting inflammation and fibrosis and improving cellular regeneration.


Subject(s)
Fibrosis , Mitochondria , Pancreatitis-Associated Proteins , Pancreatitis , Receptors, CXCR4 , Animals , Pancreatitis-Associated Proteins/metabolism , Pancreatitis-Associated Proteins/genetics , Mitochondria/metabolism , Mitochondria/pathology , Pancreatitis/pathology , Pancreatitis/metabolism , Mice , Receptors, CXCR4/metabolism , Receptors, CXCR4/genetics , Humans , Mice, Inbred C57BL , Cyclams/pharmacology , Male , Mice, Knockout , Benzylamines/pharmacology , Chemokine CXCL12/metabolism , Cell Proliferation , Signal Transduction , Autophagy , Pancreas/pathology , Pancreas/metabolism , Cell Death
9.
Int J Mol Sci ; 25(10)2024 May 17.
Article in English | MEDLINE | ID: mdl-38791525

ABSTRACT

The worldwide incidence of prediabetes/type 2 has continued to rise the last 40 years. In the same period, the mean daily energy intake has increased, and the quality of food has significantly changed. The chronic exposure of pancreatic ß-cells to calorie excess (excessive energy intake) and food additives may increase pancreatic insulin secretion, decrease insulin pulses and/or reduce hepatic insulin clearance, thereby causing chronic hyperinsulinemia and peripheral insulin resistance. Chronic calorie excess and hyperinsulinemia may promote lipogenesis, inhibit lipolysis and increase lipid storage in adipocytes. In addition, calorie excess and hyperinsulinemia can induce insulin resistance and contribute to progressive and excessive ectopic fat accumulation in the liver and pancreas by the conversion of excess calories into fat. The personal fat threshold hypothesis proposes that in susceptible individuals, excessive ectopic fat accumulation may eventually lead to hepatic insulin receptor resistance, the loss of pancreatic insulin secretion, hyperglycemia and the development of frank type 2 diabetes. Thus, type 2 diabetes seems (partly) to be caused by hyperinsulinemia-induced excess ectopic fat accumulation in the liver and pancreas. Increasing evidence further shows that interventions (hypocaloric diet and/or bariatric surgery), which remove ectopic fat in the liver and pancreas by introducing a negative energy balance, can normalize insulin secretion and glucose tolerance and induce the sustained biochemical remission of type 2 diabetes. This pathophysiological insight may have major implications and may cause a paradigm shift in the management of type 2 diabetes: avoiding/reducing ectopic fat accumulation in the liver and pancreas may both be essential to prevent and cure type 2 diabetes.


Subject(s)
Diabetes Mellitus, Type 2 , Hyperinsulinism , Overnutrition , Humans , Hyperinsulinism/metabolism , Hyperinsulinism/complications , Hyperinsulinism/etiology , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/etiology , Overnutrition/complications , Insulin Resistance , Adipose Tissue/metabolism , Animals , Liver/metabolism , Liver/pathology , Insulin/metabolism , Pancreas/metabolism , Pancreas/pathology
10.
Mol Med Rep ; 30(1)2024 07.
Article in English | MEDLINE | ID: mdl-38695254

ABSTRACT

As a pluripotent cell, activated pancreatic stellate cells (PSCs) can differentiate into various pancreatic parenchymal cells and participate in the secretion of extracellular matrix and the repair of pancreatic damage. Additionally, PSCs characteristics allow them to contribute to pancreatic inflammation and carcinogenesis. Moreover, a detailed study of the pathogenesis of activated PSCs in pancreatic disease can offer promise for the development of innovative therapeutic strategies and improved patient prognoses. Therefore, the present study review aimed to examine the involvement of activated PSCs in pancreatic diseases and elucidate the underlying mechanisms to provide a viable therapeutic strategy for the management of pancreas­related diseases.


Subject(s)
Pancreas , Pancreatic Diseases , Pancreatic Stellate Cells , Humans , Pancreatic Stellate Cells/metabolism , Pancreatic Stellate Cells/pathology , Pancreas/metabolism , Pancreas/pathology , Pancreas/cytology , Pancreatic Diseases/pathology , Pancreatic Diseases/metabolism , Animals , Extracellular Matrix/metabolism , Cell Differentiation , Pancreatic Neoplasms/pathology , Pancreatic Neoplasms/metabolism
11.
Sci Rep ; 14(1): 11141, 2024 05 15.
Article in English | MEDLINE | ID: mdl-38750103

ABSTRACT

This study aimed to analyze the characteristics of the non-specific uptake (NSU) of 18F-labeled fibroblast activation protein inhibitor (18F-FAPI) of the pancreas and investigate the related factors. Totally, 78 patients who underwent both 18F-fluorodeoxyglucose (FDG) and 18F-FAPI PET/CT examinations were divided into normal (n = 53) and NSU (n = 25) groups. The differences in general information, medical history, laboratory indexes and uptake were compared. Receiver operating characteristic (ROC) curves were used to analyze the optimal cut-off values. The correlations between 18F-FAPI-SUVmax and blood cell analysis, liver function indexes, tumor markers, and inflammatory indices were analyzed. The logistic regression model was used to estimate the independent factors. Both 18F-FAPI (4.48 ± 0.98 vs. 2.01 ± 0.53, t = 11.718, P < 0.05) and 18F-FDG (2.23 ± 0.42 vs. 2.02 ± 0.44, t = 2.036, P = 0.045) showed significantly higher in NSU group. Patients in the NSU group tended to be complicated with a history of drinking (P = 0.034), chronic liver diseases (P = 0.006), and surgery of gastrectomy (P = 0.004). ROC analysis showed cutoff values of 3.25 and 2.05 for 18F-FAPI and 18F-FDG in identifying the NSU. Patients in the NSU group showed less platelet count, higher platelet volume, higher total bilirubin, direct or indirect bilirubin (P < 0.05). Platelet count, platelet crit, large platelet ratio, aspartate aminotransferase (AST), α-L-fucosidase, and total, direct or indirect bilirubin were correlated with 18F-FAPI-SUVmax (P < 0.05). AST [1.099 (1.014, 1.192), P = 0.021] and total bilirubin [1.137 (1.035, 1.249), P = 0.007] were two independent factors in the step forward logistic regression, and platelet/% [1.079 (1.004, 1.160), P = 0.039] and total bilirubin [1.459 (1.016, 2.095), P = 0.041] were two independent factors in the step backward logistic regression for the prediction of pancreatic uptake of 18F-FAPI. 18F-FAPI-PET/CT was better than 18F-FDG in predicting the pancreatic NSU, and NSU is related to a history of drinking, chronic liver diseases, gastrectomy, heteromorphic platelet, and impaired liver function.


Subject(s)
Pancreas , Positron Emission Tomography Computed Tomography , Humans , Male , Female , Middle Aged , Pancreas/metabolism , Pancreas/diagnostic imaging , Aged , Prospective Studies , Fluorodeoxyglucose F18 , ROC Curve , Adult , Radiopharmaceuticals , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/diagnostic imaging , Pancreatic Neoplasms/surgery
12.
Pak J Pharm Sci ; 37(2): 307-314, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38767097

ABSTRACT

Long-lasting hyperglycemia can potentially cause damage to organs such as the kidneys, liver and pancreas. Glimepiride (GLIM), as a drug of choice in the treatment of diabetes mellitus (DM), has the risk of decreasing the functioning of organs such as the kidneys, liver and pancreas. Black rice bran ethanol extract (EEBRB) with antioxidant content has been shown to protect the kidney, liver and pancreas organs. The aim of this study was to establish the effect of EEBRB on lowering fasting blood glucose (FBG) and protecting several organs after GLIM administration in alloxan (ALX)-induced hyperglycemic rats. A total of 20 rats were divided into 4 groups and treated for 21 days treatments using following preparations: normal control (NC), diabetic group (DC), GLIM 1 mg/ kgBW and combination of glimepiride 1mg/kgBW and EEBRB 50 mg/KgBW (GLBR). The results showed that the GLBR was able to lower blood glucose levels back to normal (<126 mg/dL) and protect kidney, liver and pancreas cells by increasing the amount in normal cells.


Subject(s)
Blood Glucose , Diabetes Mellitus, Experimental , Hypoglycemic Agents , Kidney , Liver , Oryza , Pancreas , Plant Extracts , Sulfonylurea Compounds , Animals , Sulfonylurea Compounds/pharmacology , Plant Extracts/pharmacology , Plant Extracts/isolation & purification , Kidney/drug effects , Kidney/metabolism , Blood Glucose/drug effects , Blood Glucose/metabolism , Oryza/chemistry , Liver/drug effects , Liver/metabolism , Hypoglycemic Agents/pharmacology , Hypoglycemic Agents/isolation & purification , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Experimental/blood , Diabetes Mellitus, Experimental/chemically induced , Pancreas/drug effects , Pancreas/metabolism , Pancreas/pathology , Male , Rats , Ethanol/chemistry , Rats, Wistar
13.
Int J Mol Sci ; 25(9)2024 Apr 27.
Article in English | MEDLINE | ID: mdl-38731997

ABSTRACT

Glucose, the primary energy substrate for fetal oxidative processes and growth, is transferred from maternal to fetal circulation down a concentration gradient by placental facilitative glucose transporters. In sheep, SLC2A1 and SLC2A3 are the primary transporters available in the placental epithelium, with SLC2A3 located on the maternal-facing apical trophoblast membrane and SLC2A1 located on the fetal-facing basolateral trophoblast membrane. We have previously reported that impaired placental SLC2A3 glucose transport resulted in smaller, hypoglycemic fetuses with reduced umbilical artery insulin and glucagon concentrations, in addition to diminished pancreas weights. These findings led us to subject RNA derived from SLC2A3-RNAi (RNA interference) and NTS-RNAi (non-targeting sequence) fetal pancreases to qPCR followed by transcriptomic analysis. We identified a total of 771 differentially expressed genes (DEGs). Upregulated pathways were associated with fat digestion and absorption, particularly fatty acid transport, lipid metabolism, and cholesterol biosynthesis, suggesting a potential switch in energetic substrates due to hypoglycemia. Pathways related to molecular transport and cell signaling in addition to pathways influencing growth and metabolism of the developing pancreas were also impacted. A few genes directly related to gluconeogenesis were also differentially expressed. Our results suggest that fetal hypoglycemia during the first half of gestation impacts fetal pancreas development and function that is not limited to ß cell activity.


Subject(s)
Hypoglycemia , Pancreas , Placenta , RNA Interference , Transcriptome , Pregnancy , Animals , Female , Placenta/metabolism , Sheep , Pancreas/metabolism , Pancreas/embryology , Hypoglycemia/genetics , Hypoglycemia/metabolism , Glucose Transporter Type 3/genetics , Glucose Transporter Type 3/metabolism , Fetus/metabolism , Fetal Development/genetics , Gene Expression Regulation, Developmental , Glucose/metabolism , Gene Expression Profiling
14.
Nat Commun ; 15(1): 3744, 2024 May 03.
Article in English | MEDLINE | ID: mdl-38702321

ABSTRACT

Cellular composition and anatomical organization influence normal and aberrant organ functions. Emerging spatial single-cell proteomic assays such as Image Mass Cytometry (IMC) and Co-Detection by Indexing (CODEX) have facilitated the study of cellular composition and organization by enabling high-throughput measurement of cells and their localization directly in intact tissues. However, annotation of cell types and quantification of their relative localization in tissues remain challenging. To address these unmet needs for atlas-scale datasets like Human Pancreas Analysis Program (HPAP), we develop AnnoSpat (Annotator and Spatial Pattern Finder) that uses neural network and point process algorithms to automatically identify cell types and quantify cell-cell proximity relationships. Our study of data from IMC and CODEX shows the higher performance of AnnoSpat in rapid and accurate annotation of cell types compared to alternative approaches. Moreover, the application of AnnoSpat to type 1 diabetic, non-diabetic autoantibody-positive, and non-diabetic organ donor cohorts recapitulates known islet pathobiology and shows differential dynamics of pancreatic polypeptide (PP) cell abundance and CD8+ T cells infiltration in islets during type 1 diabetes progression.


Subject(s)
Algorithms , Diabetes Mellitus, Type 1 , Pancreas , Proteomics , Humans , Proteomics/methods , Diabetes Mellitus, Type 1/pathology , Diabetes Mellitus, Type 1/metabolism , Pancreas/cytology , Pancreas/metabolism , Islets of Langerhans/metabolism , Islets of Langerhans/cytology , Single-Cell Analysis/methods , Neural Networks, Computer , CD8-Positive T-Lymphocytes/metabolism , Image Cytometry/methods
15.
Eur J Pharmacol ; 975: 176646, 2024 Jul 15.
Article in English | MEDLINE | ID: mdl-38762157

ABSTRACT

Severe acute pancreatitis (SAP) is a complicated inflammatory reaction that impacts the pancreas, often resulting in damage to numerous organs. This disorder encompasses a range of processes such as inflammation, oxidative stress, and pancreatitis. The hormone melatonin (MT) is primarily secreted by the pineal gland and plays a crucial role in mitigating inflammation, countering the harmful effects of free radicals, and regulating oxidative stress. The aim of this research was to investigate the potential protective impact and the underlying mechanism of melatonin in mice afflicted with SAP. The biochemical and histological assessments unequivocally demonstrated that melatonin effectively inhibited necrosis, infiltration, edema and cell death in pancreatic tissues, thereby suppressing acute pancreatitis. Notably, melatonin also alleviated the consequent harm to distant organs, notably the lungs, liver, and kidneys. Furthermore, both preventive and therapeutic administration of melatonin prompted nuclear factor E2-related factor 2 (Nrf2) activation followed by Nrf2 target gene expression. Nrf2 initiates the activation of antioxidant genes, thereby providing defense against oxidative stress. Conversely, Nrf2 reduction may contribute to impaired antioxidant protection in SAP. The beneficial impact of Nrf2 on antioxidants was absent in Nrf2-knockout mice, leading to the accumulation of LDH and exacerbation of cell death. This deterioration in both pancreatitis and injuries in distant organs intensified significantly. The results indicate that melatonin has an enhanced ability to protect against multiorgan damage caused by SAP, which is accomplished through the increase in Nrf2 expression. Additionally, Nrf2 initiates the activation of antioxidant genes that offer defense against cell death.


Subject(s)
Melatonin , NF-E2-Related Factor 2 , Oxidative Stress , Pancreatitis , Signal Transduction , Animals , NF-E2-Related Factor 2/metabolism , NF-E2-Related Factor 2/genetics , Melatonin/pharmacology , Melatonin/therapeutic use , Signal Transduction/drug effects , Pancreatitis/drug therapy , Pancreatitis/pathology , Pancreatitis/metabolism , Mice , Oxidative Stress/drug effects , Male , Antioxidants/pharmacology , Antioxidants/therapeutic use , Mice, Knockout , Pancreas/drug effects , Pancreas/pathology , Pancreas/metabolism , Mice, Inbred C57BL , Acute Disease
16.
Int J Mol Sci ; 25(9)2024 Apr 26.
Article in English | MEDLINE | ID: mdl-38731942

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) can originate from acinar-to-ductal metaplasia (ADM). Pancreatic acini harboring oncogenic Kras mutations are transdifferentiated to a duct-like phenotype that further progresses to become pancreatic intraepithelial neoplasia (PanIN) lesions, giving rise to PDAC. Although ADM formation is frequently observed in KrasG12D transgenic mouse models of PDAC, the exact mechanisms of how oncogenic KrasG12D regulates this process remain an enigma. Herein, we revealed a new downstream target of oncogenic Kras, cytokine CCL9, during ADM formation. Higher levels of CCL9 and its receptors, CCR1 and CCR3, were detected in ADM regions of the pancreas in p48cre:KrasG12D mice and human PDAC patients. Knockdown of CCL9 in KrasG12D-expressed pancreatic acini reduced KrasG12D-induced ADM in a 3D organoid culture system. Moreover, exogenously added recombinant CCL9 and overexpression of CCL9 in primary pancreatic acini induced pancreatic ADM. We also showed that, functioning as a downstream target of KrasG12D, CCL9 promoted pancreatic ADM through upregulation of the intracellular levels of reactive oxygen species (ROS) and metalloproteinases (MMPs), including MMP14, MMP3 and MMP2. Blockade of MMPs via its generic inhibitor GM6001 or knockdown of specific MMP such as MMP14 and MMP3 decreased CCL9-induced pancreatic ADM. In p48cre:KrasG12D transgenic mice, blockade of CCL9 through its specific neutralizing antibody attenuated pancreatic ADM structures and PanIN lesion formation. Furthermore, it also diminished infiltrating macrophages and expression of MMP14, MMP3 and MMP2 in the ADM areas. Altogether, our results provide novel mechanistic insight into how oncogenic Kras enhances pancreatic ADM through its new downstream target molecule, CCL9, to initiate PDAC.


Subject(s)
Acinar Cells , Carcinoma, Pancreatic Ductal , Metaplasia , Pancreatic Neoplasms , Proto-Oncogene Proteins p21(ras) , Reactive Oxygen Species , Animals , Proto-Oncogene Proteins p21(ras)/genetics , Proto-Oncogene Proteins p21(ras)/metabolism , Mice , Reactive Oxygen Species/metabolism , Humans , Carcinoma, Pancreatic Ductal/metabolism , Carcinoma, Pancreatic Ductal/pathology , Carcinoma, Pancreatic Ductal/genetics , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Pancreatic Neoplasms/genetics , Metaplasia/metabolism , Metaplasia/genetics , Acinar Cells/metabolism , Acinar Cells/pathology , Mice, Transgenic , Chemokines, CC/metabolism , Chemokines, CC/genetics , Macrophage Inflammatory Proteins/metabolism , Macrophage Inflammatory Proteins/genetics , Pancreas/metabolism , Pancreas/pathology
17.
Drug Dev Res ; 85(4): e22199, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38812443

ABSTRACT

It has been reported that lipophilic statins such as atorvastatin can more readily penetrate into ß-cells and reach the mitochondria, resulting in mitochondrial dysfunction, oxidative stress, decrease in insulin release. Many studies have shown that natural products can protect mitochondrial dysfunction induced by drug in different tissue. We aimed to explore mitochondrial protection potency of hesperidin, vanillic acid, and sinapic acid as natural compounds against mitochondrial dysfunction induced by atorvastatin in pancreas isolated mitochondria. Mitochondria were isolated form rat pancreas and directly treated with toxic concentration of atorvastatin (500 µM) in presence of various concentrations hesperidin, vanillic acid, and sinapic acid (1, 10, and 100 µM) separately. Mitochondrial toxicity parameters such as the reactive oxygen species (ROS) formation, succinate dehydrogenases (SDH) activity, mitochondrial swelling, depletion of glutathione (GSH), mitochondrial membrane potential (MMP) collapse, and malondialdehyde (MDA) production were measured. Our findings demonstrated that atorvastatin directly induced mitochondrial toxicity at concentration of 500 µM and higher in pancreatic mitochondria. Except MDA, atorvastatin caused significantly reduction in SDH activity, mitochondrial swelling, ROS formation, depletion of GSH, and collapse of MMP. While, our data showed that all three protective compounds at low concentrations ameliorated atorvastatin-induced mitochondrial dysfunction with the increase of SDH activity, improvement of mitochondrial swelling, MMP collapse and mitochondrial GSH, and reduction of ROS formation. We can conclude that hesperidin, vanillic acid, and sinapic acid can directly reverse the toxic of atorvastatin in rat pancreas isolated mitochondria, which may be beneficial for protection against diabetogenic-induced mitochondrial dysfunction in pancreatic ß-cells.


Subject(s)
Atorvastatin , Coumaric Acids , Hesperidin , Membrane Potential, Mitochondrial , Mitochondria , Mitochondrial Swelling , Pancreas , Reactive Oxygen Species , Vanillic Acid , Animals , Atorvastatin/pharmacology , Mitochondria/drug effects , Mitochondria/metabolism , Pancreas/drug effects , Pancreas/pathology , Pancreas/metabolism , Coumaric Acids/pharmacology , Rats , Reactive Oxygen Species/metabolism , Male , Mitochondrial Swelling/drug effects , Membrane Potential, Mitochondrial/drug effects , Vanillic Acid/pharmacology , Hesperidin/pharmacology , Glutathione/metabolism , Rats, Wistar , Succinate Dehydrogenase/metabolism , Malondialdehyde/metabolism
18.
Cell Mol Biol (Noisy-le-grand) ; 70(5): 59-68, 2024 May 27.
Article in English | MEDLINE | ID: mdl-38814234

ABSTRACT

Development of novel functional foods is trending as one of the hot topics in food science and food/beverage industries. In the present study, the anti-diabetic, anti-hyperlipidemic and histo-protective effects of the extra virgin olive oil (EVOO) enriched with the organosulfur diallyl sulfide (DAS) (DAS-rich EVOO) were evaluated in alloxan-induced diabetic mice. The ingestion of EVOO (500µL daily for two weeks) attenuated alloxan-induced elevated glucose, alanine transaminase (ALT), aspartate transaminase (AST), alkaline phosphatase, lactate dehydrogenase (LDH), urea and creatinine. It also normalized the levels of triglycerides (TG), total cholesterols (TC), low-density lipoprotein-cholesterol (LDL-c) and their consequent atherogenic index of plasma (AIP) in diabetic animals. Additionally, EVOO prevented lipid peroxidation (MDA) and reduced the level of hydrogen peroxide (H2O2) in diabetic animals. Concomitantly, it enhanced the activity of the antioxidant enzymes catalase (CAT), glutathione peroxidase (GPx) and superoxide dismutase (SOD), reducing thereby tissue oxidative stress injury. The overall histologic (pancreas, liver, and kidney) alterations were also improved after EVOO ingestion. The manifest anti-diabetic, lipid-lowering and histo-protective properties of EVOO were markedly potentiated with DAS-rich EVOO suggesting possible synergistic interactions between DAS and EVOO lipophilic bioactive ingredients. Overall, EVOO and DAS-rich EVOO show promise as functional foods and/or adjuvants for the treatment of diabetes and its complications.


Subject(s)
Allyl Compounds , Diabetes Mellitus, Experimental , Hypoglycemic Agents , Hypolipidemic Agents , Olive Oil , Sulfides , Animals , Olive Oil/chemistry , Olive Oil/pharmacology , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Experimental/metabolism , Allyl Compounds/pharmacology , Allyl Compounds/therapeutic use , Sulfides/pharmacology , Sulfides/therapeutic use , Sulfides/chemistry , Hypoglycemic Agents/pharmacology , Hypoglycemic Agents/therapeutic use , Mice , Hypolipidemic Agents/pharmacology , Male , Antioxidants/pharmacology , Oxidative Stress/drug effects , Lipid Peroxidation/drug effects , Blood Glucose/metabolism , Blood Glucose/drug effects , Liver/drug effects , Liver/metabolism , Liver/pathology , Pancreas/drug effects , Pancreas/pathology , Pancreas/metabolism , Glutathione Peroxidase/metabolism , Catalase/metabolism , Hydrogen Peroxide/metabolism , Superoxide Dismutase/metabolism , Kidney/drug effects , Kidney/metabolism , Kidney/pathology , Alanine Transaminase/blood , Alanine Transaminase/metabolism , Aspartate Aminotransferases/metabolism , Aspartate Aminotransferases/blood , Triglycerides/blood , Triglycerides/metabolism
19.
Cell Transplant ; 33: 9636897241248942, 2024.
Article in English | MEDLINE | ID: mdl-38712762

ABSTRACT

Recently, we and others generated induced tissue-specific stem/progenitor (iTS/iTP) cells. The advantages of iTS/iTP cells compared with induced pluripotent stem (iPS) cells are (1) easier generation, (2) efficient differentiation, and (3) no teratomas formation. In this study, we generated mouse induced pancreatic stem cells (iTS-P cells) by the plasmid vector expressing Yes-associated protein 1 (YAP). The iTS-P YAP9 cells expressed Foxa2 (endoderm marker) and Pdx1 (pancreatic marker) while the expressions of Oct3/4 and Nanog (marker of embryonic stem [ES] cells) in iTS-P YAP9 cells was significantly lower compared with those in ES cells. The iTS-P YAP9 cells efficiently differentiated into insulin-expressing cells compared with ES cells. The ability to generate autologous iTS cells may be applied to diverse applications of regenerative medicine.


Subject(s)
Cell Differentiation , Induced Pluripotent Stem Cells , YAP-Signaling Proteins , Animals , Mice , Adaptor Proteins, Signal Transducing/metabolism , Adaptor Proteins, Signal Transducing/genetics , Hepatocyte Nuclear Factor 3-beta/metabolism , Hepatocyte Nuclear Factor 3-beta/genetics , Homeodomain Proteins/metabolism , Homeodomain Proteins/genetics , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Octamer Transcription Factor-3/metabolism , Octamer Transcription Factor-3/genetics , Pancreas/cytology , Pancreas/metabolism , Phosphoproteins/metabolism , Phosphoproteins/genetics , Trans-Activators/metabolism , Trans-Activators/genetics
20.
Nat Commun ; 15(1): 4528, 2024 May 29.
Article in English | MEDLINE | ID: mdl-38811532

ABSTRACT

Metabolic dysfunction-associated steatohepatitis (MASH) is the most prevalent cause of liver disease worldwide, with a single approved therapeutic. Previous research has shown that interleukin-22 (IL-22) can suppress ß-cell stress, reduce local islet inflammation, restore appropriate insulin production, reverse hyperglycemia, and ameliorate insulin resistance in preclinical models of diabetes. In clinical trials long-acting forms of IL-22 have led to increased proliferation in the skin and intestine, where the IL-22RA1 receptor is highly expressed. To maximise beneficial effects whilst reducing the risk of epithelial proliferation and cancer, we designed short-acting IL-22-bispecific biologic drugs that successfully targeted the liver and pancreas. Here we show 10-fold lower doses of these bispecific biologics exceed the beneficial effects of native IL-22 in multiple preclinical models of MASH, without off-target effects. Treatment restores glycemic control, markedly reduces hepatic steatosis, inflammation, and fibrogenesis. These short-acting IL-22-bispecific targeted biologics are a promising new therapeutic approach for MASH.


Subject(s)
Fatty Liver , Interleukin-22 , Interleukins , Liver , Pancreas , Interleukins/metabolism , Animals , Liver/metabolism , Liver/pathology , Liver/drug effects , Pancreas/pathology , Pancreas/metabolism , Pancreas/drug effects , Humans , Mice , Fatty Liver/drug therapy , Fatty Liver/metabolism , Male , Mice, Inbred C57BL , Disease Models, Animal , Insulin Resistance , Receptors, Interleukin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...