Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 3.198
Filter
1.
PLoS One ; 19(5): e0303060, 2024.
Article in English | MEDLINE | ID: mdl-38723008

ABSTRACT

In the current study we investigated the impact of combination of rutin and vitamin A on glycated products, the glyoxalase system, oxidative markers, and inflammation in animals fed a high-fat high-fructose (HFFD) diet. Thirty rats were randomly divided into six groups (n = 5). The treatments, metformin (120 mg/kg), rutin (100 mg/kg), vitamin A (43 IU/kg), and a combination of rutin (100 mg/kg) and vitamin A (43 IU/kg) were given to relevant groups of rats along with high-fructose high-fat diet for 42 days. HbA1c, D-lactate, Glyoxylase-1, Hexokinase 2, malondialdehyde (MDA), glutathione peroxidase (GPx), catalase (CAT), nuclear transcription factor-B (NF-κB), interleukin-6 (IL-6), interleukin-8 (IL-8) and histological examinations were performed after 42 days. The docking simulations were conducted using Auto Dock package. The combined effects of rutin and vitamin A in treated rats significantly (p < 0.001) reduced HbA1c, hexokinase 2, and D-lactate levels while preventing cellular damage. The combination dramatically (p < 0.001) decreased MDA, CAT, and GPx in treated rats and decreased the expression of inflammatory cytokines such as IL-6 andIL-8, as well as the transcription factor NF-κB. The molecular docking investigations revealed that rutin had a strong affinity for several important biomolecules, including as NF-κB, Catalase, MDA, IL-6, hexokinase 2, and GPx. The results propose beneficial impact of rutin and vitamin A as a convincing treatment strategy to treat AGE-related disorders, such as diabetes, autism, alzheimer's, atherosclerosis.


Subject(s)
Diet, High-Fat , Fructose , Hyperglycemia , Inflammation , Oxidative Stress , Rutin , Vitamin A , Animals , Rutin/pharmacology , Oxidative Stress/drug effects , Fructose/adverse effects , Rats , Diet, High-Fat/adverse effects , Vitamin A/pharmacology , Vitamin A/metabolism , Inflammation/metabolism , Inflammation/drug therapy , Inflammation/pathology , Male , Hyperglycemia/drug therapy , Hyperglycemia/metabolism , Hyperglycemia/chemically induced , Molecular Docking Simulation , Rats, Wistar , Disease Models, Animal , Glycosylation/drug effects , Metformin/pharmacology , Glycated Hemoglobin/metabolism , NF-kappa B/metabolism , Hexokinase/metabolism , Catalase/metabolism
2.
Front Endocrinol (Lausanne) ; 15: 1369908, 2024.
Article in English | MEDLINE | ID: mdl-38803473

ABSTRACT

Background: A phenomenon known as legacy effect was observed that poor glycemic control at early stage of patients with newly-diagnosed type 2 diabetes (T2D) increases the risk of subsequent cardiovascular diseases (CVD). Early use of some novel anti-hyperglycemic agents, such as sodium-glucose transport protein 2 inhibitors (SGLT-2i), may attenuate this effect, but the evidence is limited. Methods: Two retrospective cohorts of newly diagnosed T2D patients from 2010-2023 were assembled using the Yinzhou Regional Health Care Database (YRHCD) with different definitions of the early exposure period - the 1-year exposure cohort and 2-year exposure cohort, which were comprised of subjects who had HbA1c measurement data within 1 year and 2 years after their T2D diagnosis, respectively. Using Cox proportional hazards models, we examined the association between high HbA1c level (HbA1c>7%) during the early exposure period and the risk of subsequent CVD. This analysis was performed in the overall cohort and three subpopulations with different treatments during the early exposure period, including patients initiating SGLT-2i or glucagon-like peptide-1 receptor agonists (GLP-1RA), patients using dipeptidyl peptidase-4 inhibitors (DPP-4i), and patients without using SGLT-2i, GLP-1RA, and DPP-4i. Besides, subgroup analyses were performed by stratifying patients into age <55 and ≥55 years. Results: A total of 21,477 and 22,493 patients with newly diagnosed T2D were included in the two final cohorts. Compared with patients with mean HbA1c ≤ 7% during the early exposure period, those with HbA1c>7% had higher risks of incident CVD, with a HR of 1.165 (95%CI, 1.056-1.285) and 1.143 (95%CI, 1.044-1.252) in 1-year and 2-year exposure period cohort. Compared to non-users, in patients initiating SGLT-2i/GLP-1RA within 1 or 2 years after T2D diagnosis, higher HbA1c level at baseline was not associated with CVD in both two cohorts. In subgroup analyses, results were generally consistent with the main analysis. Conclusions: Poor glycemic control in the early stage of T2D increased later CVD risk in Chinese adults with newly diagnosed T2D. Compared to non-users, this association was smaller and non-significant in patients receiving SGLT-2i/GLP-1RA during the early stage of T2D, indicating early use of these drugs may have the potential to mitigate legacy effects of hyperglycemia.


Subject(s)
Diabetes Mellitus, Type 2 , Dipeptidyl-Peptidase IV Inhibitors , Glucagon-Like Peptide-1 Receptor , Hyperglycemia , Sodium-Glucose Transporter 2 Inhibitors , Humans , Dipeptidyl-Peptidase IV Inhibitors/therapeutic use , Dipeptidyl-Peptidase IV Inhibitors/adverse effects , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/blood , Female , Male , Middle Aged , Glucagon-Like Peptide-1 Receptor/agonists , Retrospective Studies , Sodium-Glucose Transporter 2 Inhibitors/therapeutic use , Hyperglycemia/epidemiology , Hyperglycemia/chemically induced , Aged , Cardiovascular Diseases/epidemiology , Blood Glucose/metabolism , Blood Glucose/analysis , Blood Glucose/drug effects , Glycated Hemoglobin/analysis , Glycated Hemoglobin/metabolism , Hypoglycemic Agents/therapeutic use , Adult , Follow-Up Studies , Glycemic Control , Glucagon-Like Peptide-1 Receptor Agonists
3.
J Neuroinflammation ; 21(1): 137, 2024 May 27.
Article in English | MEDLINE | ID: mdl-38802820

ABSTRACT

Hyperglycemia has been shown to modulate the immune response of peripheral immune cells and organs, but the impact of hyperglycemia on neuroinflammation within the brain remains elusive. In the present study, we provide evidences that streptozotocin (STZ)-induced hyperglycemic condition in mice drives a phenotypic switch of brain astrocytes to a proinflammatory state, and increases brain vulnerability to mild peripheral inflammation. In particular, we found that hyperglycemia led to a significant increase in the astrocyte proliferation as determined by flow cytometric and immunohistochemical analyses of mouse brain. The increased astrocyte proliferation by hyperglycemia was reduced by Glut1 inhibitor BAY-876. Transcriptomic analysis of isolated astrocytes from Aldh1l1CreERT2;tdTomato mice revealed that peripheral STZ injection induced astrocyte reprogramming into proliferative, and proinflammatory phenotype. Additionally, STZ-induced hyperglycemic condition significantly enhanced the infiltration of circulating myeloid cells into the brain and the disruption of blood-brain barrier in response to mild lipopolysaccharide (LPS) administration. Systemic hyperglycemia did not alter the intensity and sensitivity of peripheral inflammation in mice to LPS challenge, but increased the inflammatory potential of brain microglia. In line with findings from mouse experiments, a high-glucose environment intensified the LPS-triggered production of proinflammatory molecules in primary astrocyte cultures. Furthermore, hyperglycemic mice exhibited a significant impairment in cognitive function after mild LPS administration compared to normoglycemic mice as determined by novel object recognition and Y-maze tasks. Taken together, these results demonstrate that hyperglycemia directly induces astrocyte reprogramming towards a proliferative and proinflammatory phenotype, which potentiates mild LPS-triggered inflammation within brain parenchymal regions.


Subject(s)
Astrocytes , Brain , Hyperglycemia , Lipopolysaccharides , Mice, Inbred C57BL , Neuroinflammatory Diseases , Animals , Hyperglycemia/chemically induced , Hyperglycemia/pathology , Astrocytes/drug effects , Astrocytes/metabolism , Astrocytes/pathology , Mice , Lipopolysaccharides/toxicity , Lipopolysaccharides/pharmacology , Brain/pathology , Brain/metabolism , Brain/drug effects , Neuroinflammatory Diseases/metabolism , Neuroinflammatory Diseases/pathology , Neuroinflammatory Diseases/chemically induced , Male , Cellular Reprogramming/drug effects , Cellular Reprogramming/physiology , Mice, Transgenic , Cells, Cultured
4.
Ecotoxicol Environ Saf ; 279: 116494, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38820878

ABSTRACT

Di-(2-ethylhexyl)-phthalate (DEHP), as distinctive endocrine disrupting chemicals, has become a global environmental pollutant harmful to human and animal health. However, the impacts on offspring and mothers with maternal DEHP exposure are largely unknown and the mechanism remains elusive. We established DEHP-exposed maternal mice to investigate the impacts on mother and offspring and illustrate the mechanism from multiple perspectives. Pregnant mice were administered with different doses of DEHP, respectively. Metagenomic sequencing used fecal and transcriptome sequencing using placentas and livers from offspring have been performed, respectively. The results of the histopathology perspective demonstrated that DEHP exposure could disrupt the function of islets impact placentas and fetus development for maternal mice, and cause the disorder of glucose and lipid metabolism for immature offspring mice, resulting in hyperglycemia. The results of the metagenome of gut microbial communities indicated that the dysbiosis of gut microbiota in mother and offspring mice and the dominant phyla transformed through vertical transmission. Transcriptome analysis found DEHP exposure induced mutations of Ahcy and Gstp3, which can damage liver cells and affect the metabolism of the host. DEHP exposure harms pregnant mice and offspring by affecting gene expression and altering metabolism. Our results suggested that exposure of pregnant mice to DEHP during pregnancy and lactation increased the risk of metabolic disorders by altering key genes in liver and gut microbiota, and these results provided new insights into the potential long-term harms of DEHP.


Subject(s)
Diethylhexyl Phthalate , Energy Metabolism , Hyperglycemia , Maternal Exposure , Female , Animals , Pregnancy , Diethylhexyl Phthalate/toxicity , Mice , Hyperglycemia/chemically induced , Energy Metabolism/drug effects , Maternal Exposure/adverse effects , Endocrine Disruptors/toxicity , Prenatal Exposure Delayed Effects/chemically induced , Gastrointestinal Microbiome/drug effects , Environmental Pollutants/toxicity , Placenta/drug effects , Liver/drug effects
5.
Front Endocrinol (Lausanne) ; 15: 1250822, 2024.
Article in English | MEDLINE | ID: mdl-38577574

ABSTRACT

Introduction: Pasireotide, a somatostatin receptor ligand, is approved for treating acromegaly and Cushing's disease (CD). Hyperglycemia during treatment can occur because of the drug's mechanism of action, although treatment discontinuation is rarely required. The prospective, randomized, Phase IV SOM230B2219 (NCT02060383) trial was designed to assess optimal management of pasireotide-associated hyperglycemia. Here, we investigated predictive factors for requiring antihyperglycemic medication during pasireotide treatment. Methods: Participants with acromegaly or CD initiated long-acting pasireotide 40 mg/28 days intramuscularly (acromegaly) or pasireotide 600 µg subcutaneously twice daily during pre-randomization (≤16 weeks). Those who did not need antihyperglycemic medication, were managed with metformin, or received insulin from baseline entered an observational arm ending at 16 weeks. Those who required additional/alternative antihyperglycemic medication to metformin were randomized to incretin-based therapy or insulin for an additional 16 weeks. Logistic-regression analyses evaluated quantitative and qualitative factors for requiring antihyperglycemic medication during pre-randomization. Results: Of 190 participants with acromegaly and 59 with CD, 88 and 15, respectively, did not need antihyperglycemic medication; most were aged <40 years (acromegaly 62.5%, CD 86.7%), with baseline glycated hemoglobin (HbA1c) <6.5% (<48 mmol/mol; acromegaly 98.9%, CD 100%) and fasting plasma glucose (FPG) <100 mg/dL (<5.6 mmol/L; acromegaly 76.1%, CD 100%). By logistic regression, increasing baseline HbA1c (odds ratio [OR] 3.6; P=0.0162) and FPG (OR 1.0; P=0.0472) and history of diabetes/pre-diabetes (OR 3.0; P=0.0221) predicted receipt of antihyperglycemic medication in acromegaly participants; increasing baseline HbA1c (OR 12.6; P=0.0276) was also predictive in CD participants. Investigator-reported hyperglycemia-related adverse events were recorded in 47.9% and 54.2% of acromegaly and CD participants, respectively, mainly those with diabetes/pre-diabetes. Conclusion: Increasing age, HbA1c, and FPG and pre-diabetes/diabetes were associated with increased likelihood of requiring antihyperglycemic medication during pasireotide treatment. These risk factors may be used to identify those who need more vigilant monitoring to optimize outcomes during pasireotide treatment.


Subject(s)
Acromegaly , Diabetes Mellitus , Hyperglycemia , Metformin , Pituitary ACTH Hypersecretion , Prediabetic State , Somatostatin/analogs & derivatives , Humans , Acromegaly/complications , Acromegaly/drug therapy , Blood Glucose , Prediabetic State/drug therapy , Pituitary ACTH Hypersecretion/complications , Pituitary ACTH Hypersecretion/drug therapy , Prospective Studies , Hyperglycemia/chemically induced , Hyperglycemia/drug therapy , Hypoglycemic Agents/therapeutic use , Diabetes Mellitus/drug therapy , Insulin/therapeutic use , Metformin/therapeutic use
6.
BMJ Case Rep ; 17(4)2024 Apr 02.
Article in English | MEDLINE | ID: mdl-38569738

ABSTRACT

A man in his late 60s with a history of well-controlled type 2 diabetes and hepatic cirrhosis presented to the emergency department due to uncontrollable hyperglycaemia following the initial brentuximab vedotin (BV) infusion. BV was initiated as a treatment for mycosis fungoides, a form of cutaneous T-cell lymphoma. The patient was diagnosed with severe hyperglycaemia with ketosis. Empiric treatment with amoxicillin-clavulanic acid, hydration and intravenous insulin infusion was initiated. Hyperglycaemia persisted despite receiving massive amounts of insulin and was corrected only after treatment with high-dose methylprednisolone for suspected type B insulin resistance. Extremely high and difficult-to-treat hyperglycaemia is a rare side effect of BV. Unfortunately, the patient died of upper gastrointestinal bleeding 22 days after discharge. In patients with obesity and/or diabetes mellitus, the blood glucose levels should be carefully monitored when treated with BV.


Subject(s)
Diabetes Mellitus, Type 2 , Hyperglycemia , Immunoconjugates , Insulin Resistance , Insulins , Skin Neoplasms , Male , Humans , Brentuximab Vedotin/therapeutic use , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/drug therapy , Skin Neoplasms/pathology , Hyperglycemia/chemically induced , Hyperglycemia/drug therapy , Insulins/therapeutic use
7.
Neurology ; 102(9): e209323, 2024 May 14.
Article in English | MEDLINE | ID: mdl-38626363

ABSTRACT

BACKGROUND AND OBJECTIVES: Baseline hyperglycemia is associated with worse outcomes in acute ischemic stroke (AIS), including higher risk of symptomatic intracerebral hemorrhage (sICH) following treatment with thrombolysis. Prospective data are lacking to inform management of post-thrombolysis hyperglycemia. In a prespecified analysis from the Stroke Hyperglycemia Insulin Network Effort (SHINE) trial of hyperglycemic stroke management, we hypothesized that post-thrombolysis hyperglycemia is associated with a higher risk of sICH. METHODS: Hyperglycemic AIS patients <12 hours onset were randomized to intensive insulin (target range 80-130 mg/dL) vs standard sliding scale (80-179 mg/dL) over a 72-hour period, stratified by treatment with thrombolysis. Three board-certified vascular neurologists independently reviewed all sICH events occurring within 7 days, defined by neurologic deterioration of ≥4 points on the NIH Stroke Scale (NIHSS). Associations between blood glucose control and sICH were analyzed using logistic regression accounting for NIHSS, age, systolic blood pressure, onset to thrombolysis time, and endovascular therapy (odds ratios [OR], 95% CI). Additional analysis compared patients in a high-risk group (age older than 60 years and NIHSS ≥8) vs all others. Categorical variables and outcomes were compared using the χ2 test (p < 0.05). RESULTS: Of 1151 SHINE participants, 725 (63%) received thrombolysis (median age 65 years, 46% women, 29% Black, 18% Hispanic). The median NIHSS was 7, baseline blood glucose was 187 (interquartile range 153-247) mg/dL, and 80% were diabetic. Onset to thrombolysis time was 2.2 hours (1.6-2.9). Post-thrombolysis sICH occurred in 3.6% (3.0% intensive vs 4.3% standard glucose control, OR 1.10, 0.60-2.01, p = 0.697). In the first 12 hours, every 10 mg/dL higher glucose increased the odds of sICH (OR 1.08, 1.03-1.14, p = 0.004), and a greater proportion of glucose measures in the normal range (80-130 mg/dL) decreased the odds of sICH (0.89, 0.80-0.99, p = 0.030). These associations were strongest in the high-risk group (age older than 60 years and NIHSS ≥8). DISCUSSION: In this prespecified analysis from the SHINE trial, intensive insulin therapy was not associated with a reduced risk of post-thrombolysis sICH compared with standard sliding scale. However, early post-thrombolysis hyperglycemia was associated with a higher risk of sICH overall, particularly in older patients with more severe strokes. Further prospective research is warranted to address the risk of sICH in hyperglycemic stroke patients undergoing endovascular therapy. TRIAL REGISTRATION INFORMATION: NCT01369069.


Subject(s)
Brain Ischemia , Hyperglycemia , Insulins , Ischemic Stroke , Stroke , Humans , Female , Aged , Middle Aged , Male , Tissue Plasminogen Activator/adverse effects , Blood Glucose , Fibrinolytic Agents/adverse effects , Stroke/complications , Stroke/drug therapy , Ischemic Stroke/drug therapy , Brain Ischemia/complications , Brain Ischemia/drug therapy , Thrombolytic Therapy/adverse effects , Treatment Outcome , Cerebral Hemorrhage/chemically induced , Cerebral Hemorrhage/epidemiology , Cerebral Hemorrhage/complications , Hyperglycemia/chemically induced , Hyperglycemia/complications , Hyperglycemia/drug therapy , Insulins/therapeutic use
8.
Toxicol Ind Health ; 40(5): 232-243, 2024 May.
Article in English | MEDLINE | ID: mdl-38467557

ABSTRACT

Exposure of zebrafish embryos to glucose is a suitable model for the fetal hyperglycemia seen in gestational diabetes. Diethylhexyl phthalate (DEHP), which is considered an endocrine-disrupting chemical, is one of the most common phthalate derivatives used in stretching plastic and is encountered in every area where plastic is used in daily life. In the present study, the effects of DEHP on pathways related to insulin resistance and obesity were examined in zebrafish embryos exposed to glucose as a fetal hyperglycemia model. Zebrafish embryos were exposed to DEHP, glucose, and glucose + DEHP for 72 h post-fertilization (hpf), and developmental parameters and locomotor activities were monitored. At 72 hpf ins, lepa, pparγ, atf4a, and il-6 expressions were determined by RT-PCR. Glucose, lipid peroxidation (LPO), nitric oxide (NO) levels, glutathione S-transferase (GST), superoxide dismutase (SOD), and acetylcholine esterase (AChE) activities were measured spectrophotometrically. Compared with the control group, glucose, LPO, GST activity, il6, and atf4a expressions increased in all exposure groups, while body length, locomotor, and SOD activities decreased. While AChE activity decreased in the DEHP and glucose groups, it increased in the glucose + DEHP group. Although glucose exposure increased pparγ and lepa expressions, DEHP significantly decreased the expressions of pparγ and lepa both in the DEHP and glucose + DEHP groups. Our findings showed that DEHP amplified oxidant and inflammatory responses in this fetal hyperglycemia model, predisposing insulin resistance in zebrafish embryos.


Subject(s)
Diethylhexyl Phthalate , Hyperglycemia , Insulin Resistance , Animals , Diethylhexyl Phthalate/toxicity , Zebrafish/metabolism , Oxidants , PPAR gamma , Glucose/metabolism , Hyperglycemia/chemically induced , Superoxide Dismutase
9.
Clin Pharmacol Ther ; 115(6): 1408-1417, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38425181

ABSTRACT

Thiazide diuretics, widely used in hypertension, cause a variety of adverse reactions, including hyperglycemia, hyperuricemia, and electrolyte abnormalities. In this study, we aimed to identify genetic variants that interact with thiazide-use to increase the risk of these adverse reactions. Using UK Biobank data, we first performed genomewide variance quantitative trait locus (vQTL) analysis of ~ 6.2 million SNPs on 95,493 unrelated hypertensive White British participants (24,313 on self-reported bendroflumethiazide treatment at recruitment) for 2 blood (glucose and urate) and 2 urine (potassium and sodium) biomarkers. Second, we conducted direct gene-environment interaction (GEI) tests on the significant (P < 2.5 × 10-9) vQTLs, included a second UK Biobank cohort comprising 13,647 unrelated hypertensive White British participants (3,478 on thiazides other than bendroflumethiazide) and set significance at P = 0.05 divided by the number of vQTL SNPs tested for GEIs. The vQTL analysis identified eight statistically significant SNPs for blood glucose (5 SNPs) and serum urate (3 SNPs), with none being identified for the urinary biomarkers. Two of the SNPs (1 glucose SNP: CDKAL1 intron rs35612982, GEI P = 6.24 × 10-3; and 1 serum urate SNP: SLC2A9 intron rs938564, GEI P = 4.51 × 10-4) demonstrated significant GEI effects in the first, but not the second, cohort. Both genes are biologically plausible candidates, with the SLC2A9-mediated interaction having been previously reported. In conclusion, we used a two-stage approach to detect two biologically plausible genetic loci that can interact with thiazides to increase the risk of thiazide-associated biochemical abnormalities. Understanding how environmental exposures (including medications such as thiazides) and genetics interact, is an important step toward precision medicine and improved patient outcomes.


Subject(s)
Biological Specimen Banks , Genome-Wide Association Study , Hyperglycemia , Hyperuricemia , Polymorphism, Single Nucleotide , Sodium Chloride Symporter Inhibitors , Humans , United Kingdom/epidemiology , Female , Hyperuricemia/genetics , Hyperuricemia/urine , Hyperuricemia/chemically induced , Male , Middle Aged , Hyperglycemia/genetics , Hyperglycemia/chemically induced , Hyperglycemia/urine , Hyperglycemia/epidemiology , Aged , Sodium Chloride Symporter Inhibitors/adverse effects , Uric Acid/urine , Uric Acid/blood , Quantitative Trait Loci , Gene-Environment Interaction , Hypertension/genetics , Hypertension/chemically induced , Blood Glucose/drug effects , Blood Glucose/metabolism , Potassium/urine , Potassium/blood , Sodium/urine , Adult , Biomarkers/urine , Biomarkers/blood , UK Biobank
10.
Breast Cancer Res ; 26(1): 36, 2024 03 04.
Article in English | MEDLINE | ID: mdl-38439079

ABSTRACT

BACKGROUND: Hyperglycemia is an on-target effect of PI3Kα inhibitors. Early identification and intervention of treatment-induced hyperglycemia is important for improving management of patients receiving a PI3Kα inhibitor like alpelisib. Here, we characterize incidence of grade 3/4 alpelisib-related hyperglycemia, along with time to event, management, and outcomes using a machine learning model. METHODS: Data for the risk model were pooled from patients receiving alpelisib ± fulvestrant in the open-label, phase 1 X2101 trial and the randomized, double-blind, phase 3 SOLAR-1 trial. The pooled population (n = 505) included patients with advanced solid tumors (X2101, n = 221) or HR+/HER2- advanced breast cancer (SOLAR-1, n = 284). External validation was performed using BYLieve trial patient data (n = 340). Hyperglycemia incidence and management were analyzed for SOLAR-1. RESULTS: A random forest model identified 5 baseline characteristics most associated with risk of developing grade 3/4 hyperglycemia (fasting plasma glucose, body mass index, HbA1c, monocytes, age). This model was used to derive a score to classify patients as high or low risk for developing grade 3/4 hyperglycemia. Applying the model to patients treated with alpelisib and fulvestrant in SOLAR-1 showed higher incidence of hyperglycemia (all grade and grade 3/4), increased use of antihyperglycemic medications, and more discontinuations due to hyperglycemia (16.7% vs. 2.6% of discontinuations) in the high- versus low-risk group. Among patients in SOLAR-1 (alpelisib + fulvestrant arm) with PIK3CA mutations, median progression-free survival was similar between the high- and low-risk groups (11.0 vs. 10.9 months). For external validation, the model was applied to the BYLieve trial, for which successful classification into high- and low-risk groups with shorter time to grade 3/4 hyperglycemia in the high-risk group was observed. CONCLUSIONS: A risk model using 5 clinically relevant baseline characteristics was able to identify patients at higher or lower probability for developing alpelisib-induced hyperglycemia. Early identification of patients who may be at higher risk for hyperglycemia may improve management (including monitoring and early intervention) and potentially lead to improved outcomes. REGISTRATION: ClinicalTrials.gov: NCT01219699 (registration date: October 13, 2010; retrospectively registered), ClinicalTrials.gov: NCT02437318 (registration date: May 7, 2015); ClinicalTrials.gov: NCT03056755 (registration date: February 17, 2017).


Subject(s)
Breast Neoplasms , Hyperglycemia , Thiazoles , Humans , Female , Breast Neoplasms/drug therapy , Fulvestrant/adverse effects , Hyperglycemia/chemically induced , Hyperglycemia/epidemiology , Risk Assessment
11.
Endocrinol Metab (Seoul) ; 39(2): 222-238, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38532282

ABSTRACT

Glucocorticoids provide a potent therapeutic response and are widely used to treat a variety of diseases, including coronavirus disease 2019 (COVID-19) infection. However, the issue of glucocorticoid-induced hyperglycemia (GIH), which is observed in over one-third of patients treated with glucocorticoids, is often neglected. To improve the clinical course and prognosis of diseases that necessitate glucocorticoid therapy, proper management of GIH is essential. The key pathophysiology of GIH includes systemic insulin resistance, which exacerbates hepatic steatosis and visceral obesity, as well as proteolysis and lipolysis of muscle and adipose tissue, coupled with ß-cell dysfunction. For patients on glucocorticoid therapy, risk stratification should be conducted through a detailed baseline evaluation, and frequent glucose monitoring is recommended to detect the onset of GIH, particularly in high-risk individuals. Patients with confirmed GIH who require treatment should follow an insulin-centered regimen that varies depending on whether they are inpatients or outpatients, as well as the type and dosage of glucocorticoid used. The ideal strategy to maintain normoglycemia while preventing hypoglycemia is to combine basal-bolus insulin and correction doses with a continuous glucose monitoring system. This review focuses on the current understanding and latest evidence concerning GIH, incorporating insights gained from the COVID-19 pandemic.


Subject(s)
COVID-19 , Glucocorticoids , Hyperglycemia , Humans , Glucocorticoids/adverse effects , Glucocorticoids/therapeutic use , Glucocorticoids/administration & dosage , Hyperglycemia/chemically induced , SARS-CoV-2 , Blood Glucose/analysis , Blood Glucose/drug effects , Insulin/administration & dosage , Insulin Resistance , COVID-19 Drug Treatment
12.
Endocr Pract ; 30(4): 367-371, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38307456

ABSTRACT

OBJECTIVE: There is a relative lack of consensus regarding the optimal management of hyperglycemia in patients receiving continuous enteral nutrition (EN), with or without a diagnosis of diabetes. METHODS: This retrospective study examined 475 patients (303 with known diabetes) hospitalized in critical care setting units in 2019 in a single center who received continuous EN. Rates of hypoglycemia, hyperglycemia, and glucose levels within the target range (70-180 mg/dL) were compared between patients with and without diabetes, and among patients treated with intermediate-acting (IA) biphasic neutral protamine Hagedorn 70/30, long-acting (LA) insulin, or rapid-acting insulin only. RESULTS: Among those with type 2 diabetes mellitus, IA and LA insulin regimens were associated with a significantly higher proportion of patient-days in the target glucose range and fewer hyperglycemic days. Level 1 (<70 mg/dL) and level 2 (<54 mg/dL) hypoglycemia occurred rarely, and there were no significant differences in level 2 hypoglycemia frequency across the different insulin regimens. CONCLUSION: Administration of IA and LA insulin can be safe and effective for those receiving insulin doses for EN-related hyperglycemia.


Subject(s)
Diabetes Mellitus, Type 2 , Hyperglycemia , Hypoglycemia , Humans , Diabetes Mellitus, Type 2/drug therapy , Hypoglycemic Agents/adverse effects , Retrospective Studies , Enteral Nutrition , Critical Illness/therapy , Blood Glucose , Insulin/adverse effects , Hypoglycemia/chemically induced , Hypoglycemia/epidemiology , Hypoglycemia/drug therapy , Insulin, Long-Acting/therapeutic use , Hyperglycemia/drug therapy , Hyperglycemia/prevention & control , Hyperglycemia/chemically induced , Glucose/therapeutic use , Insulin, Isophane/adverse effects
13.
Tohoku J Exp Med ; 262(4): 263-268, 2024 May 01.
Article in English | MEDLINE | ID: mdl-38325830

ABSTRACT

Anamorelin (ANAM) is a novel ghrelin receptor agonist for the treatment of cancer cachexia. In clinical trials of ANAM, glucose metabolism disorders as adverse effects were relatively frequent, however, when and how they occur remains unclear. Moreover, the safety in patients with pancreatic cancer and/or diabetes has not been clarified because most previous studies focused on patients with non-small cell lung cancer and had excluded patients with poorly controlled diabetes. Herein, a 66-year-old man with advanced pancreatic cancer and diabetes was administered ANAM, and acute hyperglycemia was developed and could be monitored by the self-monitoring of blood glucose (SMBG). Increasing the insulin dose failed to control hyperglycemia adequately, but the hyperglycemia ameliorated quickly after ANAM discontinuation. The continuous glucose monitoring (CGM) revealed that the sensor glucose levels had remained in the high range throughout the day during ANAM administration despite using 1.5 times more insulin. Our report is one of the few that describe the details of ANAM-induced hyperglycemia and provides important information for the safe and effective use of ANAM.


Subject(s)
Hyperglycemia , Pancreatic Neoplasms , Humans , Male , Hyperglycemia/chemically induced , Hyperglycemia/drug therapy , Hyperglycemia/complications , Aged , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/complications , Diabetes Mellitus/drug therapy , Oligopeptides/adverse effects , Oligopeptides/therapeutic use , Blood Glucose , Hydrazines/adverse effects , Hydrazines/therapeutic use , Neoplasm Staging , Acute Disease
14.
Article in English | MEDLINE | ID: mdl-38311294

ABSTRACT

Birds have the highest blood glucose among vertebrates. Several mechanisms may explain this including the lack of a functional insulin-responsive glucose transport protein, high glucagon concentrations, and reliance on lipid oxidation resulting in the production of gluconeogenic precursors. The hypothesis was that interruption of gluconeogenesis using the diabetes medication metformin would lower glucose concentrations in wild-caught birds. We captured two cohorts of adult mourning doves, Zenaida macroura, and acclimated them to captivity for two weeks. In this crossover study, cohort 1 was administered a single dose of one of the following oral treatments each week: metformin (150 or 300 mg/kg), glycogenolysis inhibitor (2.5 mg/kg 1,4-dideoxy-1,4-imino-D-arabinitol (DAB)), or water (50 µL). Whole blood glucose was measured using a glucometer at baseline, 30, 60, and 120 min following the oral doses. In contrast to mammals and chickens, 300 mg/kg metformin did not alter blood glucose (p > 0.05) whereas 150 mg/kg metformin increased blood glucose compared to water (p = 0.043). To examine whether 150 mg/kg metformin stimulated glycogenolysis, we co-administered 150 mg/kg metformin and 2.5 mg/kg DAB, which prevented the hyperglycemic response. Cohort 2 was administered the same treatments and the early response was examined (0, 5, 10, 15 min). Low-dose metformin increased blood glucose within 5 min (p = 0.039) whereas the high dose had no effect. DAB did not prevent the early response to metformin nor did it alter blood glucose concentrations when administered alone (p = 0.887). In conclusion, metformin increases endogenous blood glucose via glycogenolysis in healthy adult male mourning doves.


Subject(s)
Hyperglycemia , Metformin , Humans , Male , Animals , Columbidae , Blood Glucose , Metformin/pharmacology , Cross-Over Studies , Chickens , Hyperglycemia/chemically induced , Animals, Wild , Water , Grief , Mammals
15.
Acta Med Port ; 37(3): 207-214, 2024 Feb 05.
Article in Portuguese | MEDLINE | ID: mdl-38316163

ABSTRACT

Hyperglycaemia affects more than 30% of adults hospitalized for non-critical illness and is associated with an increased risk of adverse clinical outcomes. Insulin therapy is widely used for its safety and efficacy. However, given the growing availability of new drugs and new classes of antidiabetic agents with benefits beyond glycaemic control, challenges arise regarding their use in the hospital setting. This article aims to review and summarize the most recently available evidence and recommendations on the role of non-insulin antidiabetic agents in the management of hyperglycaemia in hospitalized patients. Insulin therapy remains the method of choice. Dipeptidyl peptidase 4 inhibitors can be considered in mild to moderate hyperglycaemia. Glucagon-like peptide 1 receptor agonists have recently shown promising results, with high efficacy in glycaemic control and low risk of hypoglycaemia. There are concerns regarding the increased risk of acidosis with metformin use, especially in cases of acute illness, although there is no evidence to support its suspension in selected patients with relative clinical stability. Sodium-glucose cotransporter-2 inhibitors should be discontinued in clinical situations that may predispose to ketoacidosis, including episodes of acute illness. The hospital use of sulfonylureas and thiazolidinediones is not advised.


A hiperglicemia afeta mais de 30% dos adultos hospitalizados por doença não crítica e está associada a um risco aumentado de desfechos clínicos adversos. A insulinoterapia é amplamente utilizada pela sua segurança e eficácia. Contudo, face à disponibilidade crescente de novos fármacos antidiabéticos com benefícios além do controlo glicémico, surgem desafios quanto à sua utilização em contexto hospitalar. Este artigo tem como objetivo rever e sumariar a evidência e as recomendações mais recentemente disponibilizadas sobre o papel dos antidiabéticos não insulínicos na gestão da hiperglicemia a nível hospitalar. A insulinoterapia mantém-se como o método de eleição. Os inibidores da dipeptidil peptidase 4 podem ser considerados em casos de hiperglicemia ligeira a moderada, como alternativa ou de forma complementar à insulinoterapia. Os agonistas dos recetores do glucagon-like peptide 1 têm recentemente revelado resultados promissores, com elevada eficácia no controlo glicémico e risco baixo de hipoglicemia. Existem preocupações relativas ao risco acrescido de acidose com a metformina, sobretudo em casos de doença aguda, apesar de não existir evidência que suporte a sua suspensão em doentes selecionados e com relativa estabilidade clínica. Os inibidores do cotransportador de sódio-glicose-2 devem ser descontinuados em situações clínicas que possam predispor a cetoacidose, incluindo episódios de doença aguda. A utilização hospitalar das sulfonilureias e das tiazolidinedionas é desaconselhada.


Subject(s)
Diabetes Mellitus, Type 2 , Hyperglycemia , Sodium-Glucose Transporter 2 Inhibitors , Adult , Humans , Hypoglycemic Agents/adverse effects , Insulin/therapeutic use , Hyperglycemia/chemically induced , Hyperglycemia/drug therapy , Sodium-Glucose Transporter 2 Inhibitors/therapeutic use , Acute Disease , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/drug therapy
16.
Gut Microbes ; 16(1): 2310277, 2024.
Article in English | MEDLINE | ID: mdl-38332701

ABSTRACT

Up to 40% of transplant recipients treated long-term with tacrolimus (TAC) develop post-transplant diabetes mellitus (PTDM). TAC is an important risk factor for PTDM, but is also essential for immunosuppression after transplantation. Long-term TAC treatment alters the gut microbiome, but the mechanisms of TAC-induced gut microbiota in the pathogenesis of PTDM are poorly characterized. Here, we showed that vancomycin, an inhibitor of bacterial beta-glucuronidase (GUS), prevents TAC-induced glucose disorder and insulin resistance in mice. Metagenomics shows that GUS-producing bacteria are predominant and flourish in the TAC-induced hyperglycemia mouse model, with upregulation of intestinal GUS activity. Targeted metabolomics analysis revealed that in the presence of high GUS activity, the hydrolysis of bile acid (BAs)-glucuronic conjugates is increased and most BAs are overproduced in the serum and liver, which, in turn, activates the ileal farnesoid X receptor (FXR) and suppresses GLP-1 secretion by L-cells. The GUS inhibitor vancomycin significantly eliminated GUS-producing bacteria and inhibited bacterial GUS activity and BAs levels, thereby enhancing L-cell GLP-1 secretion and preventing hyperglycemia. Our results propose a novel clinical strategy for inhibiting the bacterial GUS enzyme to prevent hyperglycemia without requiring withdrawal of TAC treatment. This strategy exerted its effect through the ileal bile acid-FXR-GLP-1 pathway.


Subject(s)
Diabetes Mellitus , Gastrointestinal Microbiome , Hyperglycemia , Mice , Animals , Tacrolimus/pharmacology , Tacrolimus/therapeutic use , Vancomycin/pharmacology , Immunosuppressive Agents/therapeutic use , Hyperglycemia/chemically induced , Hyperglycemia/drug therapy , Bacteria/genetics , Bacteria/metabolism , Glucuronidase/metabolism , Glucuronidase/pharmacology , Bile Acids and Salts/pharmacology , Glucagon-Like Peptide 1
17.
Can J Diabetes ; 48(3): 195-203.e1, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38211830

ABSTRACT

OBJECTIVES: The aim of this study was to determine the associations between glucocorticoid administration during chemotherapy for hematologic malignancy and hyperglycemia, new-onset diabetes, and mortality in Ontario, Canada. Hospitalization and emergency room utilization during the chemotherapy treatment period were also described. METHODS: We conducted a retrospective cohort study using health administrative data from ICES, Ontario, to assess risk of new-onset diabetes, new-onset hyperglycemia, and hyperglycemia for individuals with leukemia, non-Hodgkin lymphoma (NHL), and Hodgkin lymphoma (HL) receiving glucocorticoids during chemotherapy between 2006 and 2016. Using multivariable regression models, we determined the associations between glucocorticoid exposure and our outcomes of interest, controlling for age, sex, marginalization, and comorbidities. RESULTS: Our cohort included 19,530 individuals; 71.1% (n=13,893) received a glucocorticoid. The highest proportion of hyperglycemia occurred with leukemia (25.4%, n=1,301). Of the 15,580 individuals with no history of diabetes, those with leukemia had the highest rate of new-onset diabetes (7.1%, n=279) and new-onset hyperglycemia (18.1%, n=641), and glucocorticoid exposure increased the risk of new-onset diabetes (hazard ratio [HR] 1.29, 95% confidence interval [CI] 1.01 to 1.64, p=0.04) and new-onset hyperglycemia (HR 1.28, 95% CI 1.09 to 1.5, p=0.003). Hyperglycemia during chemotherapy increased the risk of all-cause mortality for the combined (HR 1.18, 95% CI 1.09 to 1.27, p<0.0001) and NHL (HR 1.16, 95% CI 1.04 to 1.28, p=0.007) cohorts. CONCLUSIONS: Hyperglycemia is common during hematologic chemotherapy treatment and is associated with a modest increased risk of all-cause mortality. Routine screening, monitoring, and management of hyperglycemia should be an integral part of treatment plans for leukemia, NHL, or HL, with or without glucocorticoid administration.


Subject(s)
Diabetes Mellitus , Glucocorticoids , Hematologic Neoplasms , Hyperglycemia , Humans , Female , Hyperglycemia/epidemiology , Hyperglycemia/chemically induced , Hyperglycemia/mortality , Male , Glucocorticoids/adverse effects , Glucocorticoids/therapeutic use , Middle Aged , Retrospective Studies , Diabetes Mellitus/epidemiology , Diabetes Mellitus/mortality , Aged , Hematologic Neoplasms/mortality , Hematologic Neoplasms/drug therapy , Hematologic Neoplasms/epidemiology , Adult , Cohort Studies , Ontario/epidemiology , Antineoplastic Agents/adverse effects , Antineoplastic Agents/therapeutic use , Lymphoma, Non-Hodgkin/drug therapy , Lymphoma, Non-Hodgkin/mortality , Lymphoma, Non-Hodgkin/epidemiology
18.
J Pediatr Endocrinol Metab ; 37(4): 367-370, 2024 Apr 25.
Article in English | MEDLINE | ID: mdl-38281148

ABSTRACT

OBJECTIVES: Hyperglycemia is a known side effect of anticancer chemotherapeutic drugs. This entity known as drug-induced diabetes mellitus usually does not present with the development of diabetic ketoacidosis (DKA). We hereby report a case of drug induced diabetes mellitus in a child with acute leukemia presenting with DKA. CASE PRESENTATION: We report a case of a teenage boy diagnosed with B cell acute lymphoblastic leukemia and was started on induction phase chemotherapy as per the Indian Collaborative Childhood Leukemia group (ICICLe) acute lymphoblastic leukemia-14 protocol. On day 12 of the induction phase, he developed hyperglycemia and presented to us with severe diabetic ketoacidosis (DKA). Serum anti glutamic acid decarboxylase 65 antibody levels were negative with low serum C peptide levels. Initially, the possibility of drug-induced acute pancreatitis was kept which was ruled out. Keeping the possibility of drug-induced hyperglycemia, the child was started on subcutaneous regular insulin which was titrated as per sugar records. Continuation of remaining chemotherapy was done by PEGylated L-asparaginase with titration of insulin as per home-based sugar records. Insulin requirement increased from 0.3 unit/kg/day to a maximum of 1 unit/kg/day during consolidation phase 1 with PEGylated L-asparaginase suggesting drug-induced hyperglycemia but subsequently insulin requirement decreased and insulin was stopped. CONCLUSIONS: Drug induced diabetes mellitus can present as DKA during induction phase of acute lymphoblastic leukemia (ALL) chemotherapy. A high index of suspicion and close monitoring are required. The insulin requirements in these patients can be very fluctuant and may become nil during the course of treatment.


Subject(s)
Antineoplastic Agents , Diabetes Mellitus , Diabetic Ketoacidosis , Hyperglycemia , Pancreatitis , Precursor Cell Lymphoblastic Leukemia-Lymphoma , Adolescent , Humans , Male , Acute Disease , Antineoplastic Agents/adverse effects , Asparaginase/adverse effects , Diabetes Mellitus/drug therapy , Diabetic Ketoacidosis/diagnosis , Hyperglycemia/chemically induced , Insulin/therapeutic use , Pancreatitis/chemically induced , Pancreatitis/complications , Pancreatitis/drug therapy , Polyethylene Glycols/adverse effects , Precursor Cell Lymphoblastic Leukemia-Lymphoma/complications , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Sugars/adverse effects
19.
Pharmacol Rep ; 76(1): 185-194, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38273183

ABSTRACT

BACKGROUND: Graves' orbitopathy (GO) is an autoimmune disorder of the orbit and retro-ocular tissues and the primary extrathyroidal manifestation of Graves' disease. In moderate-to-severe and active GO iv glucocorticoids (GCs) are recommended as first-line treatment. The aim was to assess the safety profile of methylprednisolone administered intravenously for three consecutive days at 1 g in patients with active, moderate-to-severe or sight-threatening Graves' orbitopathy. METHODS: We retrospectively evaluated 161 medical records of patients with GO treated with high-dose systemic GCs in the Department of Endocrinology, Metabolic Disorders, and Internal Medicine in Poznan between 2014 and 2021. Clinical data included age, gender, laboratory results, activity and severity of GO, smoking status, disease duration, and presented side effects. RESULTS: The presence of mild side effects was observed during 114 (71%) hospitalizations. The most common complications were hyperglycemia (n = 95) and elevated aminotransferases (n = 31). Increased levels of aminotransferases were more likely observed in smokers and GO duration above 12 months. Based on the multivariate logistic regression, higher TRAb and CAS values were significantly associated with lower odds of hyperglycemia. In turn, the increased odds of elevated aminotransferases were significantly correlated with higher initial ALT levels, female gender, and GO duration above 12 months. In addition, the multidimensional correspondence analysis (MPA) showed that GO patients who declared smoking and had not L-ornithine L-aspartate applied demonstrated a higher probability of elevated aminotransferases. CONCLUSIONS: Active GO treatment with high-dose systemic GCs is not associated with serious side effects. Hyperglycemia is the most common steroid-induced complication.


Subject(s)
Graves Disease , Graves Ophthalmopathy , Hyperglycemia , Humans , Female , Graves Ophthalmopathy/drug therapy , Graves Ophthalmopathy/etiology , Retrospective Studies , Graves Disease/complications , Graves Disease/drug therapy , Glucocorticoids/adverse effects , Methylprednisolone/adverse effects , Hyperglycemia/chemically induced , Hyperglycemia/drug therapy , Transaminases
20.
PLoS One ; 19(1): e0297809, 2024.
Article in English | MEDLINE | ID: mdl-38295137

ABSTRACT

BACKGROUND: The stress hyperglycemia ratio (SHR), a newly developed metric, is used to assess adverse outcomes in patients with acute ischemic stroke (AIS). However, the relationship between SHR and fatal outcomes (in-hospital mortality [IHM], malignant cerebral edema [MCE], symptomatic intracerebral hemorrhage [sICH], 3-month mortality, and poor functional outcome) in AIS patients receiving recombinant tissue plasminogen activator (rt-PA) treatment is unclear, and determining the optimal threshold remains incomplete. MATERIALS AND METHODS: We retrospectively enrolled a total of 345 AIS patients treated with rt-PA during 2015-2022 and collected data on various glucose metrics, including different types of SHR, glycemic gap (GG), random plasma glucose (RPG), fasting plasma glucose (FPG), and hemoglobin A1c (HbA1c). SHR and GG were calculated using these equations: SHR1, [FPG]/[HbA1c]; SHR2, [admission RPG]/[HbA1c]; SHR3, FPG/[(1.59 × HbA1c)-2.59]; SHR4, [admission RPG]/[(1.59 × HbA1c)-2.59]; GG, admission RPG - [(1.59 × HbA1c)-2.59]. We used multivariable logistic regression analysis (MVLR) to identify the association between different glucose metrics and outcomes while comparing their predictive values. RESULTS: SHR1 had the greatest predictive power and a more significant correlation with fatal outcomes than other continuous glucose metrics. The area under the curve of the SHR1 for IHM, MCE, and sICH, 3-month mortality, and poor functional outcome were 0.75, 0.77, 0.77, 0.76, and 0.73, respectively. SHR1 (per 1-point increases) was independently associated with IHM (Odds ratios [ORs] = 5.80; 95% CI [1.96, 17.17]; p = 0.001), MCE (ORs = 4.73; 95% CI [1.71, 13.04]; p = 0.003), sICH (ORs = 4.68, 95% CI [1.48-14.82]; p = 0.009), 3-month mortality (ORs = 10.87; 95% CI [3.56, 33.21]; p<0.001), and 3-month poor functional outcome (ORs = 8.05; 95% CI [2.77, 23.39]; p<0.001) after adjustment in MVLR. In subgroup analysis, elevated SHR1 was associated with fatal outcomes in patients with non-diabetes, SBP≥ 180 mmHg, and NIHSS <16. CONCLUSION: SHR1 demonstrates an independent association with fatal outcomes in AIS patients treated with rt-PA, exhibiting superior predictive ability over other glucose metrics.


Subject(s)
Brain Ischemia , Hyperglycemia , Ischemic Stroke , Stroke , Humans , Tissue Plasminogen Activator/therapeutic use , Ischemic Stroke/complications , Blood Glucose , Glycated Hemoglobin , Retrospective Studies , Treatment Outcome , Risk Factors , Hyperglycemia/complications , Hyperglycemia/chemically induced , Cerebral Hemorrhage/complications , Glucose , Brain Ischemia/drug therapy , Brain Ischemia/complications
SELECTION OF CITATIONS
SEARCH DETAIL
...