Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 36
Filter
1.
J Endocrinol ; 260(1)2024 01 01.
Article in English | MEDLINE | ID: mdl-37855335

ABSTRACT

Reports in North America suggest that up to 20% of young women (18-24 years) use cannabis during pregnancy. This is concerning given clinical studies indicate that maternal cannabis use is associated with fetal growth restriction and dysglycemia in the offspring. Preclinical studies demonstrated that prenatal exposure to Δ9-tetrahydrocannabinol, the main psychoactive component of cannabis, in rat dams led to female-specific deficits in ß-cell mass and glucose intolerance/insulin resistance. Yet to date, the contributions of cannabidiol (CBD), the primary nonpsychoactive compound in cannabis, remain elusive. This study aimed to define the effects of in utero cannabidiol (CBD) exposure on postnatal glucose regulation. Pregnant Wistar rat dams received daily intraperitoneal injections of either a vehicle solution or 3 mg/kg of CBD from gestational day (GD) 6 to parturition. CBD exposure did not lead to observable changes in maternal or neonatal outcomes; however, by 3 months of age male CBD-exposed offspring exhibited glucose intolerance despite no changes in pancreatic ß/α-cell mass. Transcriptomic analysis on the livers of these CBD-exposed males revealed altered gene expression of circadian rhythm clock machinery, which is linked to systemic glucose intolerance. Furthermore, alterations in hepatic developmental and metabolic processes were also observed, suggesting gestational CBD exposure has a long-lasting detrimental effect on liver health throughout life. Collectively, these results indicate that exposure to CBD alone in pregnancy may be detrimental to the metabolic health of the offspring later in life.


Subject(s)
Cannabidiol , Glucose Intolerance , Insulin Resistance , Insulin-Secreting Cells , Pregnancy , Rats , Female , Male , Humans , Animals , Infant , Cannabidiol/toxicity , Glucose Intolerance/chemically induced , Rats, Wistar
2.
Can J Diabetes ; 46(8): 851-862, 2022 Dec.
Article in English | MEDLINE | ID: mdl-35985923

ABSTRACT

OBJECTIVES: Administration of Δ9-tetrahydrocannabinol (Δ9-THC) to pregnant rats results in glucose intolerance, insulin resistance and reduced islet mass in female, but not male, offspring. The effects of Δ9-THC on other islet hormones is not known. One downstream target of the cannabinoid receptor, stathmin-2 (Stmn2), has recently been shown to suppress glucagon secretion, thereby suggesting Δ9-THC may also affect alpha-cell function. The aim of the present study was to determine the effects of in-utero Δ9-THC exposure on the profile of glucagon, insulin and Stmn2 in the rat offspring islet and serum. METHODS: Pregnant Wistar rat dams were injected with Δ9-THC (3 mg/kg per day, intraperitoneally) or vehicle from gestational day 6 to birth. Offspring were euthanized at postnatal day 21 (PND21) or at 5 months (adult) to collect blood and pancreata. RESULTS: At PND21, control and Δ9-THC-exposed offspring showed that Stmn2 had a strong colocalization with glucagon (Pearson's correlation coefficient ≥0.6), and a weak colocalization with insulin (Pearson's correlation coefficient <0.4) in both males and females, with no changes by either treatment or sex. In adult female offspring in the Δ9-THC group, intensity analysis indicated an increased insulin-to-glucagon (I/G; p<0.05) ratio and a decreased glucagon-to-Stmn2 (G/S; p<0.01) ratio, and no changes in these ratios in adult males. Furthermore, Δ9-THC did not alter fasting blood glucose and serum insulin levels in either male or female adult offspring. However, female Δ9-THC-exposed offspring exhibited an increased I/G ratio (p<0.05) and decreased G/S ratio in serum by adulthood (p<0.05). CONCLUSION: Collectively, the reduced G/S ratio in both islet and serum in association with an increased serum I/G ratio has direct correlations with early glucose intolerance and insulin resistance observed exclusively in females' offspring in this prenatal cannabinoid model.


Subject(s)
Glucose Intolerance , Insulin Resistance , Prenatal Exposure Delayed Effects , Animals , Female , Male , Pregnancy , Rats , Dronabinol/adverse effects , Glucagon , Insulin , Prenatal Exposure Delayed Effects/chemically induced , Rats, Wistar , Stathmin
3.
Int J Mol Sci ; 22(23)2021 Nov 29.
Article in English | MEDLINE | ID: mdl-34884717

ABSTRACT

Gestational diabetes mellitus results, in part, from a sub-optimal ß-cell mass (BCM) during pregnancy. Artemisinins were reported to increase BCM in models of diabetes by α- to ß-cell conversion leading to enhanced glucose tolerance. We used a mouse model of gestational glucose intolerance to compare the effects of an artemisinin (artesunate) on glycemia of pregnant mice with vehicle treatment (acetone) or no treatment. Animals were treated daily from gestational days (GD) 0.5 to 6.5. An intraperitoneal glucose tolerance test was performed prior to euthanasia at GD18.5 or post-partum. Glucose tolerance was significantly improved in both pregnant and non-pregnant mice with both artesunate and vehicle-alone treatment, suggesting the outcome was primarily due to the acetone vehicle. In non-pregnant, acetone-treated animals, improved glucose tolerance was associated with a higher BCM and a significant increase in bihormonal insulin and glucagon-containing pancreatic islet cells, suggesting α- to ß-cell conversion. BCM did not differ with treatment during pregnancy or post-partum. However, placental weight was higher in acetone-treated animals and was associated with an upregulation of apelinergic genes. Acetone-treated animals had reduced weight gain during treatment despite comparable food consumption to non-treated mice, suggesting transient effects on nutrient uptake. The mean duodenal and ileum villus height was reduced following exposure to acetone. We conclude that acetone treatment may mimic transient fasting, resulting in a subsequent improvement in glucose tolerance during pregnancy.


Subject(s)
Acetone/pharmacology , Antimalarials/therapeutic use , Artesunate/therapeutic use , Diabetes, Gestational/drug therapy , Pancreas/drug effects , Animals , Apelin/metabolism , Disease Models, Animal , Drug Evaluation, Preclinical , Fasting , Female , Intestines/drug effects , Placenta/drug effects , Placenta/metabolism , Pregnancy , Pregnancy Outcome
4.
J Dev Orig Health Dis ; 12(6): 915-922, 2021 12.
Article in English | MEDLINE | ID: mdl-33353580

ABSTRACT

Although abundant evidence exists that adverse events during pregnancy lead to chronic conditions, there is limited information on the impact of acute insults such as sepsis. This study tested the hypothesis that impaired fetal development leads to altered organ responses to a septic insult in both male and female adult offspring. Fetal growth restricted (FGR) rats were generated using a maternal protein-restricted diet. Male and female FGR and control diet rats were housed until 150-160 d of age when they were exposed either a saline (control) or a fecal slurry intraperitoneal (Sepsis) injection. After 6 h, livers and lungs were analyzed for inflammation and, additionally, the amounts and function of pulmonary surfactant were measured. The results showed increases in the steady-state mRNA levels of inflammatory cytokines in the liver in response to the septic insult in both males and females; these responses were not different between FGR and control diet groups. In the lungs, cytokines were not detectable in any of the experimental groups. A significant decrease in the relative amount of surfactant was observed in male FGR offspring, but this was not observed in control males or in female animals. Overall, it is concluded that FGR induced by maternal protein restriction does not impact liver and lung inflammatory response to sepsis in either male or female adult rats. An altered septic response in male FGR offspring with respect to surfactant may imply a contribution to lung dysfunction.


Subject(s)
Diet, Protein-Restricted/adverse effects , Sepsis/physiopathology , Animal Nutritional Physiological Phenomena , Animals , Diet, Protein-Restricted/methods , Disease Models, Animal , Quebec , Rats , Rats, Wistar , Sepsis/diet therapy
5.
J Comp Neurol ; 529(2): 421-433, 2021 02.
Article in English | MEDLINE | ID: mdl-32447764

ABSTRACT

Combination of ischemia and ß-amyloid (Aß) toxicity has been shown to simultaneously increase neuro-inflammation, endogenous Aß deposition, and neurodegeneration. However, studies on the evolution of infarct and panorama of cellular degeneration as a synergistic or overlapping mechanism between ischemia and Aß toxicity are lacking. Here, we compared fluorojade B (FJB) and hematoxylin and eosin (H&E) stains primarily to examine the chronology of infarct, and the viability and morphological changes in neuroglia and neurons located in different brain regions on d1, d7, and d28 post Aß toxicity and endothelin-1 induced ischemia (ET1) in rats. We demonstrated a regional difference in cellular degeneration between cortex, corpus callosum, striatum, globus pallidus, and thalamus after cerebral injury. Glial cells in the cortex and corpus callosum underwent delayed FJB staining from d7 to d28, but neurons in cortex disappeared within the first week of cerebral injury. Striatal lesion core and globus pallidus of Aß + ET1 rats showed extensive degeneration of neuronal cells compared with ET1 rats alone starting from d1. Differential and exacerbated expressions of cyclooxygenase-2 might be the cause of excessive neuronal demise in the striatum of Aß + ET1 rats. Such an investigation may improve our understanding to identify and manipulate a critical therapeutic window post comorbid injury.


Subject(s)
Brain Injuries/pathology , Cell Survival/physiology , Cerebral Cortex/pathology , Cerebral Cortex/physiology , Nerve Degeneration/pathology , Animals , Cerebral Cortex/chemistry , Corpus Striatum/chemistry , Corpus Striatum/pathology , Corpus Striatum/physiology , Male , Rats , Rats, Wistar
6.
J Endocrinol ; 246(2): 175-187, 2020 08.
Article in English | MEDLINE | ID: mdl-32485676

ABSTRACT

Maternal diabetes impairs fetal development and increases the risk of metabolic diseases in the offspring. Previously, we demonstrated that maternal dietary supplementation with 6% of olive oil prevents diabetes-induced embryo and fetal defects, in part, through the activation of peroxisome proliferator-activated receptors (PPARs). In this study, we examined the effects of this diet on neonatal and adult pancreatic development in male and female offspring of mothers affected with pre-gestational diabetes. A mild diabetic model was developed by injecting neonatal rats with streptozotocin (90 mg/kg). During pregnancy, these dams were fed a chow diet supplemented or not with 6% olive oil. Offspring pancreata was examined at day 2 and 5 months of age by immunohistochemistry followed by morphometric analysis to determine number of islets, α and ß cell clusters and ß-cell mass. At 5 months, male offspring of diabetic mothers had reduced ß-cell mass that was prevented by maternal supplementation with olive oil. PPARα and PPARγ were localized mainly in α cells and PPARß/δ in both α and ß cells. Although Pparß/δ and Pparγ RNA expression showed reduction in 5-month-old male offspring of diabetic rats, Pparß/δ expression returned to control levels after olive-oil supplementation. Interestingly, in vitro exposure to oleic acid (major component of olive oil) and natural PPAR agonists such as LTB4, CPC and 15dPGJ2 also significantly increased expression of all Ppars in αTC1-6 cells. However, only oleic acid and 15dPGJ2 increased insulin and Pdx-1 expression in INS-1E cells suggesting a protective role in ß-cells. Olive oil may be considered a dietary supplement to improve islet function in offspring of affected mothers with pre-gestational diabetes.


Subject(s)
Diabetes Mellitus, Experimental/diet therapy , Diabetes, Gestational/diet therapy , Olive Oil/therapeutic use , Animals , Dietary Supplements , Female , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Leukotriene B4/pharmacology , Lipid Metabolism/drug effects , Male , Oleic Acid/therapeutic use , Peroxisome Proliferator-Activated Receptors/agonists , Pregnancy , Rats , Streptozocin/toxicity , Trans-Activators/genetics , Trans-Activators/metabolism
7.
Reprod Toxicol ; 94: 84-91, 2020 06.
Article in English | MEDLINE | ID: mdl-32325173

ABSTRACT

Recent reports indicate that 7% of pregnant mothers in North America use cannabis. This is concerning given that in utero exposure to Δ9-tetrahydrocannabinol (Δ9-THC), the main psychoactive component in cannabis, causes fetal growth restriction and may alter replication and survival of pancreatic ß-cells in the offspring. Accordingly, we hypothesized that maternal exposure to Δ9-THC during pregnancy would impair postnatal glucometabolic health of offspring. To test this hypothesis, pregnant Wistar rats were treated with daily intraperitoneal injections of either 3 mg/kg Δ9-THC or vehicle from gestational day 6 to birth. Offspring were subsequently challenged with glucose and insulin at 5 months of age to assess glucose tolerance and peripheral muscle insulin sensitivity. Female offspring exposed to Δ9-THC in utero were glucose intolerant, associated with blunted insulin response in muscle and increased serum insulin concentration 15 min after glucose challenge. Additionally, pancreata from male and female offspring were harvested at postnatal day 21 and 5 months of age for assessment of endocrine pancreas morphometry by immunostaining. This analysis revealed that gestational exposure to Δ9-THC reduced the density of islets in female, but not male, offspring at postnatal day 21 and 5 months, culminating in reduced ß-cell mass at 5 months. These results demonstrate that fetal exposure to Δ9-THC causes female-specific impairments in glucose homeostasis, raising concern regarding the metabolic health of offspring, particularly females, exposed to cannabis in utero.


Subject(s)
Dronabinol/toxicity , Glucose/metabolism , Islets of Langerhans/drug effects , Prenatal Exposure Delayed Effects , Animals , Female , Homeostasis/drug effects , Insulin/metabolism , Islets of Langerhans/growth & development , Male , Maternal-Fetal Exchange , Pregnancy , Rats, Wistar , Sex Characteristics
8.
Cell Mol Neurobiol ; 40(3): 369-380, 2020 Apr.
Article in English | MEDLINE | ID: mdl-31522299

ABSTRACT

Besides other causes, ischemia and Alzheimer's disease pathology is also linked to decreased cerebral blood flow (CBF). There is little or no consensus about the role of neuroglial cells in maintaining CBF in various neuropathologies. This consensus becomes scarcer when it comes to clinical and experimental cases of comorbid Abeta-amyloid (Aß) toxicity and ischemia. Here, a comorbid rat model of Aß toxicity and endothelin-1 induced ischemia (ET1) not only demonstrated the appearance of axotomized phagocytosed pyknotic neurons (NeuN) immediately after the injury, but also showed a diversity of continuously changing neuroglia (MHC Class II/OX6, Iba1) and macrophage (Iba1/CD68) phenotypes with round, stout somas, and retracted processes. This is indicative of a response to a concomitant increase in large fluid-filled spaces due to the vascular leakage. Ironically 4 weeks after the injury despite a conclusive reduction in neurons, CBF restoration in ET1 rats was associated with a massive increase in neuroglial cell numbers, hypertrophy, ramification, and soma sizes bordering the continuously reducing lesion core and inflamed vasculature, possibly to shield their leaky phenotype. Astrocytes were also found to be releasing matrix metalloproteinase9 (MMP9), which stabilized matrix ligand ß-dystroglycan (ß-DG) in repaired or functional vessels. Changing neuroglia phenotypes, responses, motility, astrocytic recruitment of MMP9, and ß-DG stabilization implies the role of communication between neuroglia and endothelium in recovering CBF, in the absence of neurons, in ET1 rats compared to Aß+ET1 rats, which showed characteristics delayed neuroglial activation. Stimulation of timely neuroglial reactivity may serve as a viable strategy to compensate for the neuronal loss in restoring CBF in comorbid cases of ischemia and Aß toxicity.


Subject(s)
Alzheimer Disease/complications , Alzheimer Disease/pathology , Brain Ischemia/complications , Brain Ischemia/pathology , Cerebrovascular Circulation/physiology , Neuroglia/physiology , Alzheimer Disease/chemically induced , Alzheimer Disease/physiopathology , Amyloid beta-Peptides , Animals , Astrocytes/pathology , Astrocytes/physiology , Brain/blood supply , Brain/pathology , Brain/physiopathology , Brain Ischemia/physiopathology , Disease Models, Animal , Male , Neurons/pathology , Neurons/physiology , Rats , Rats, Wistar , Reperfusion Injury/complications , Reperfusion Injury/pathology , Reperfusion Injury/physiopathology
9.
J Comp Neurol ; 528(1): 48-60, 2020 01 01.
Article in English | MEDLINE | ID: mdl-31265125

ABSTRACT

Large cerebral infarctions are major predictors of death and severe disability from stroke. Conversely, data concerning these types of infarctions and the affected adjacent brain circuits are scarce. It remains to be determined if the co-morbid concurrence of large infarct and ß-amyloid (Aß) toxicity can precipitate the early development of dementia. Here, we described a dose-dependent effect of a unilateral striatal injection of vasoconstrictive endothelin-1 (ET-1) along with Aß toxicity on CNS pathogenesis; driven by the anatomical and functional networks within a brain circuit. After 21 days of treatment, a high dose (60 pmol) of ET-1 (E60) alone caused the greatest increase in neuroinflammation, mainly in the ipsilateral striatum and distant regions with synaptic links to the striatal lesion such as white matter (subcortical white matter, corpus callosum, internal capsule, anterior commissure), gray matter (globus pallidus, thalamus), and cortices (cingulate, motor, somatosensory, entorhinal). The combined E60 + Aß treatment also extended perturbation in the contralateral hemisphere of these rats, such as increased deposition of amyloid precursor protein fragments associated with the appearance of degenerating cells and the leakage of laminin from the basement membrane across a compromised blood-brain barrier. However, the cerebral damage induced by the 6 pmol ET-1 (E6), Aß and E6 + Aß rats was not detrimental enough to injure the complete network. The appreciation of the causal interactions among distinct anatomical units in the brain after ischemia and Aß toxicity will help in the design of effective and alternative therapeutics that may disassociate the synergistic or additive association between the infarcts and Aß toxicity.


Subject(s)
Amyloid beta-Peptides/toxicity , Brain/pathology , Cerebral Infarction/pathology , Endothelin-1/toxicity , Nerve Net/pathology , Amyloid beta-Peptides/administration & dosage , Animals , Brain/anatomy & histology , Brain/drug effects , Cerebral Infarction/chemically induced , Endothelin-1/administration & dosage , Injections, Intraventricular , Male , Nerve Net/anatomy & histology , Nerve Net/drug effects , Rats , Rats, Wistar
10.
Cell Mol Neurobiol ; 40(3): 381-382, 2020 04.
Article in English | MEDLINE | ID: mdl-31599387

ABSTRACT

The original version of this article contained a random order of part labels for Fig. 4. The correct caption of Fig. 4 with correct order of part labels is given below.

11.
Mol Neurobiol ; 56(4): 2714-2727, 2019 Apr.
Article in English | MEDLINE | ID: mdl-30054859

ABSTRACT

The hippocampus, a brain region vital for memory and learning, is sensitive to the damage caused by ischemic/hypoxic stroke and is one of the main regions affected by Alzheimer's disease. The pathological changes that might occur in the hippocampus and its connections, because of cerebral injury in a distant brain region, such as the striatum, have not been examined. Therefore, in the present study, we evaluated the combined effects of endothelin-1-induced ischemia (ET1) in the striatum and ß-amyloid (Aß) toxicity on hippocampal pathogenesis, dictated by the anatomical and functional intra- and inter-regional hippocampal connections to the striatum. The hippocampal pathogenesis induced by Aß or ET1 alone was not severe enough to significantly affect the entire circuit of the hippocampal network. However, the combination of the two pathological states (ET1 + Aß) led to an exacerbated increase in neuroinflammation, deposition of the amyloid precursor protein (APP) fragments with the associated appearance of degenerating cells, and blood-brain-barrier disruption. This was observed mainly in the hippocampal formation (CA2 and CA3 regions), the dentate gyrus as well as distinct regions with synaptic links to the hippocampus such as entorhinal cortex, thalamus, and basal forebrain. In addition, ET1 + Aß-treated rats also demonstrated protracted loss of AQP4 depolarization, dissolution of ß-dystroglycan, and basement membrane laminin with associated IgG and dysferlin leakage. Spatial dynamics of hippocampal injury in ET1 + Aß rats may provide a valuable model to study new targets for clinical therapeutic applications, specifically when areas remotely connected to hippocampus are damaged.


Subject(s)
Amyloid beta-Peptides/toxicity , Corpus Striatum/pathology , Hippocampus/blood supply , Hippocampus/injuries , Hippocampus/pathology , Animals , Corpus Striatum/drug effects , Hippocampus/metabolism , Male , Rats, Wistar
12.
J Biol Chem ; 293(44): 17218-17228, 2018 11 02.
Article in English | MEDLINE | ID: mdl-30217817

ABSTRACT

Fibroblast growth factor receptor-1 (FGFR1) activity at the plasma membrane is tightly controlled by the availability of co-receptors and competing receptor isoforms. We have previously shown that FGFR1 activity in pancreatic beta-cells modulates a wide range of processes, including lipid metabolism, insulin processing, and cell survival. More recently, we have revealed that co-expression of FGFR5, a receptor isoform that lacks a tyrosine-kinase domain, influences FGFR1 responses. We therefore hypothesized that FGFR5 is a co-receptor to FGFR1 that modulates responses to ligands by forming a receptor heterocomplex with FGFR1. We first show here increased FGFR5 expression in the pancreatic islets of nonobese diabetic (NOD) mice and also in mouse and human islets treated with proinflammatory cytokines. Using siRNA knockdown, we further report that FGFR5 and FGFR1 expression improves beta-cell survival. Co-immunoprecipitation and quantitative live-cell imaging to measure the molecular interaction between FGFR5 and FGFR1 revealed that FGFR5 forms a mixture of ligand-independent homodimers (∼25%) and homotrimers (∼75%) at the plasma membrane. Interestingly, co-expressed FGFR5 and FGFR1 formed heterocomplexes with a 2:1 ratio and subsequently responded to FGF2 by forming FGFR5/FGFR1 signaling complexes with a 4:2 ratio. Taken together, our findings identify FGFR5 as a co-receptor that is up-regulated by inflammation and promotes FGFR1-induced survival, insights that reveal a potential target for intervention during beta-cell pathogenesis.


Subject(s)
Cytokines/immunology , Diabetes Mellitus/genetics , Insulin-Secreting Cells/immunology , Receptor, Fibroblast Growth Factor, Type 1/genetics , Receptor, Fibroblast Growth Factor, Type 5/genetics , Animals , Diabetes Mellitus/immunology , Dimerization , Female , Fibroblast Growth Factor 2/immunology , Humans , Mice , Mice, Inbred BALB C , Mice, Inbred NOD , Receptor, Fibroblast Growth Factor, Type 1/chemistry , Receptor, Fibroblast Growth Factor, Type 1/immunology , Receptor, Fibroblast Growth Factor, Type 5/chemistry , Receptor, Fibroblast Growth Factor, Type 5/immunology , Up-Regulation
13.
Islets ; 10(4): 137-150, 2018.
Article in English | MEDLINE | ID: mdl-30110202

ABSTRACT

Both bone marrow-derived hematopoietic stem cells (HSC) and mesenchymal stem cells (MSC) improve glycemic control in diabetic mice, but their kinetics and associated changes in pancreatic morphology have not been directly compared. Our goal was to examine the time course of improvements in glucose tolerance and associated changes in ß-cell mass and proliferation following transplantation of equivalent numbers of HSC or MSC from the same bone marrow into diabetic non-obese diabetic severe combined immune deficiency (NOD.SCID) mice. We used transgenic mice with a targeted expression of yellow fluorescent protein (YFP) driven by the Vav1 gene promoter to genetically tag HSC and progeny. HSC were separated from bone marrow by fluorescence-activated cell sorting and MSC following cell culture. Equivalent numbers of isolated HSC or MSC were transplanted directly into the pancreas of NOD.SCID mice previously made diabetic with streptozotocin. Glucose tolerance, serum insulin, ß-cell mass and ß-cell proliferation were examined up to 28 days following transplant. Transplantation with MSC improved glucose tolerance within 7 days and serum insulin levels increased, but with no increase in ß-cell mass. Mice transplanted with HSC showed improved glucose tolerance only after 3 weeks associated with increased ß-cell proliferation and mass. We conclude that single injections of either MSC or HSC transiently improved glycemic control in diabetic NOD.SCID mice, but with different time courses. However, only HSC infiltrated the islets and were associated with an expanded ß-cell mass. This suggests that MSC and HSC have differing mechanisms of action.


Subject(s)
Hematopoietic Stem Cells/physiology , Hyperglycemia/therapy , Insulin/blood , Mesenchymal Stem Cells/physiology , Stem Cell Transplantation/methods , Animals , Blood Glucose/metabolism , Bone Marrow , Disease Models, Animal , Glucose Tolerance Test/methods , Insulin-Secreting Cells/pathology , Insulin-Secreting Cells/physiology , Mice , Mice, Inbred NOD , Mice, SCID , Time Factors , Treatment Outcome
14.
Sci Rep ; 8(1): 5136, 2018 03 23.
Article in English | MEDLINE | ID: mdl-29572520

ABSTRACT

Ischemic stroke and diabetes are vascular risk factors for the development of impaired memory such as dementia and/or Alzheimer's disease. Clinical studies have demonstrated that minor striatal ischemic lesions in combination with ß-amyloid (Aß) load are critical in generating cognitive deficits. These cognitive deficits are likely to be associated with impaired insulin signaling. In this study, we examined the histological presence of insulin-like growth factor-I (IGF-1) and insulin receptor substrate (IRS-1) in anatomically distinct brain circuits compared with morphological brain damage in a co-morbid rat model of striatal ischemia (ET1) and Aß toxicity. The results demonstrated a rapid increase in the presence of IGF-1 and IRS-1 immunoreactive cells in Aß + ET1 rats, mainly in the ipsilateral striatum and distant regions with synaptic links to the striatal lesion. These regions included subcortical white matter, motor cortex, thalamus, dentate gyrus, septohippocampal nucleus, periventricular region and horizontal diagonal band of Broca in the basal forebrain. The alteration in IGF-1 and IRS-1 presence induced by ET1 or Aß rats alone was not severe enough to affect the entire brain circuit. Understanding the causal or etiologic interaction between insulin and IGF signaling and co-morbidity after ischemia and Aß toxicity will help design more effective therapeutics.


Subject(s)
Amyloid beta-Peptides/metabolism , Brain Ischemia/metabolism , Cognition Disorders/metabolism , Corpus Striatum/metabolism , Insulin-Like Growth Factor I/metabolism , Insulin/metabolism , Memory Disorders/metabolism , Signal Transduction , Animals , Brain Ischemia/pathology , Cognition Disorders/pathology , Comorbidity , Corpus Striatum/pathology , Disease Models, Animal , Insulin Receptor Substrate Proteins/metabolism , Male , Memory Disorders/pathology , Rats , Rats, Wistar
15.
J Endocr Soc ; 2(2): 178-189, 2018 Feb 01.
Article in English | MEDLINE | ID: mdl-29450407

ABSTRACT

Ghrelin and its receptor, the growth hormone secretagogue receptor 1a (GHSR1a), are present in cardiac tissue. Activation of GHSR1a by ghrelin promotes cardiomyocyte contractility and survival, and changes in myocardial GHSR1a and circulating ghrelin track with end-stage heart failure, leading to the hypothesis that GHSR1a is a biomarker for heart failure. We hypothesized that GHSR1a could also be a biomarker for diabetic cardiomyopathy (DCM). We used two models of streptozotocin (STZ)-induced DCM: group 1, adult mice treated with 35 mg/kg STZ for 3 days; and group 2, neonatal mice treated with 70 mg/kg STZ at days 2 and 5 after birth. In group 1, mild fasting hyperglycemia (11 mM) was first detected 8 weeks after the last injection, and in group 2, severe fasting hyperglycemia (20 mM) was first detected 1 to 3 weeks after the last injection. In group 1, left ventricular function was slightly impaired as measured by echocardiography, and Western blot analysis showed a significant decrease in myocardial GHSR1a. In group 2, GHSR1a levels were also decreased as assessed by Cy5-ghrelin(1-19) fluorescence microscopy, and there was a significant negative correlation between GHSR1a levels and glucose tolerance. There were significant positive correlations between GHSR1a and ghrelin and between GHSR1a and sarcoplasmic reticulum Ca2+-ATPase 2a (SERCA2a), a marker for contractility, but not between GHSR1a and B-type natriuretic peptide, a marker for heart failure. We conclude that the subclinical stage of DCM is accompanied by alterations in the myocardial ghrelin-GHSR1a system, suggesting the possibility of a biomarker for DCM.

16.
Islets ; 8(3): 65-82, 2016 04 18.
Article in English | MEDLINE | ID: mdl-27010375

ABSTRACT

Regeneration of insulin-producing ß-cells from resident pancreas progenitors requires an understanding of both progenitor identity and lineage plasticity. One model suggested that a rare ß-cell sub-population within islets demonstrated multi-lineage plasticity. We hypothesized that ß-cells from young mice (postnatal day 7, P7) exhibit such plasticity and used a model of islet dedifferentiation toward a ductal epithelial-cell phenotype to test this theory. RIPCre;Z/AP(+/+) mice were used to lineage trace the fate of ß-cells during dedifferentiation culture by a human placental alkaline phosphatase (HPAP) reporter. There was a significant loss of HPAP-expressing ß-cells in culture, but remaining HPAP(+) cells lost insulin expression while gaining expression of the epithelial duct cell marker cytokeratin-19 (Ck19). Flow cytometry and recovery of ß-cell subpopulations from whole pancreas vs. islets suggest that the HPAP(+)Ck19(+) cells had derived from insulin-positive, glucose-transporter-2-low (Ins(+)Glut2(LO)) cells, representing 3.5% of all insulin-expressing cells. The majority of these cells were found outside of islets within clusters of <5 ß-cells. These insulin(+)Glut2(LO) cells demonstrated a greater proliferation rate in vivo and in vitro as compared to insulin(+)Glut2(+) cells at P7, were retained into adulthood, and a subset differentiated into endocrine, ductal, and neural lineages, illustrating substantial plasticity. Results were confirmed using RIPCre;ROSA- eYFP mice. Quantitative PCR data indicated these cells possess an immature ß-cell phenotype. These Ins(+)Glut2(LO) cells may represent a resident population of cells capable of forming new, functional ß-cells, and which may be potentially exploited for regenerative therapies in the future.


Subject(s)
Cell Plasticity , Glucose Transport Proteins, Facilitative/analysis , Insulin-Secreting Cells/physiology , Insulin/analysis , Animals , Cell Differentiation , Cell Lineage/physiology , Flow Cytometry , Keratin-19/analysis , Mice , Microscopy, Fluorescence
17.
Horm Mol Biol Clin Investig ; 25(3): 171-9, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26812801

ABSTRACT

BACKGROUND: Exposure to bisphenol A (BPA), an endocrine disrupting chemical, during gestation is associated with a variety of metabolic dysfunctions in adulthood, including hyperinsulinemia, glucose intolerance and insulin resistance. These modifications in glucose homeostasis largely stem from alterations in pancreatic function. However, the effects of BPA on the fetal pancreas have never been explored. The present study addressed this important question by examining the effects of prenatal BPA exposure on the mouse fetal pancreatic development. MATERIALS AND METHODS: Pregnant mice were fed a BPA diet (25 mg BPA/kg diet) from embryonic day 7.5 (E7.5) to E18.5. At E18.5, fetal pancreata were collected and analyzed for morphological changes in the endocrine pancreas such as islet size, number and ß and α cell distribution. RESULTS: We showed that BPA exposed fetal pancreata had a greater number of islet-cell clusters (ICCs; <300 µm(2); p<0.05) compared with controls. Furthermore, immunohistochemical analysis revealed that prenatal BPA exposure increased both glucagon expression in islets and the numbers of glucagon-expressing islet-cell clusters (p<0.05). CONCLUSION: Considering that ICCs represent the initial stages of islet development in the fetal pancreas, our findings suggest that BPA promotes islet differentiation or delays the conversion of ICCs into mature islets. Moreover, the increase in glucagon expression suggests a potential alteration in the α:ß-cell ratio in islets, which may have significant implications for the fetal pancreas both structurally and functionally. This study provides novel insight into the effects of BPA exposure on the fetal pancreata, indicating alterations in glucagon expression in islets and ICCs.


Subject(s)
Benzhydryl Compounds/adverse effects , Endocrine Disruptors/adverse effects , Islets of Langerhans/drug effects , Islets of Langerhans/embryology , Phenols/adverse effects , Prenatal Exposure Delayed Effects/chemically induced , Animals , Female , Glucagon-Secreting Cells/cytology , Glucagon-Secreting Cells/drug effects , Glucagon-Secreting Cells/pathology , Insulin-Secreting Cells/cytology , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/pathology , Islets of Langerhans/pathology , Islets of Langerhans/ultrastructure , Mice , Mice, Inbred C57BL , Pregnancy , Prenatal Exposure Delayed Effects/pathology
18.
Reproduction ; 149(3): 293-303, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25667428

ABSTRACT

The study carried out in our laboratory demonstrated that protein restriction (low protein, LP) during fetal and neonatal life alters pancreatic development and impairs glucose tolerance later in life. In this study, we examined the role of the transcription factor Pdx1, a master regulator of ß-cell differentiation and function along with its downstream target genes insulin, Glut2 and glucokinase (GK). The role(s) of these genes and protein products on the pancreata of male offspring from mothers exposed to LP diets were assessed during gestation, weaning, and adult life. Pregnant rats were allocated to two dietary treatments: control (C) 20% protein diet or LP, 8% protein diet. At birth, offspring were divided into four groups: C received control diet all life, LP1 received LP diet all life, LP2 changed the LP diet to C at weaning, and LP3 switched to C after being exposed to LP during gestation only. Body weights (bw) were significantly (P<0.001) decreased in all LP groups at birth. At weaning, only the LP3 offspring had their body weight restored to control levels. Pdx1 or any of the Pdx1-target genes were similar in all diets at day 21. However, at d130 Pdx1 mRNA expression and protein abundance were significantly decreased (P<0.05) in all LP groups. In addition, insulin mRNA and protein were decreased in LP1 and LP3 groups compared with C, Glut2 mRNA and GLUT2 protein levels were decreased in LP3 and GK did not change between groups. Intraperitoneal glucose tolerance test revealed impaired glucose tolerance in LP3 males, concomitant with decreased ß-cell mass, islet area, and PDX1 nuclear protein localization. Collectively, this study suggests that restoring proteins in the diet after birth in LP offspring dramatically impairs glucose homeostasis in early adulthood, by altering Pdx1 expression and downstream-target genes increasing the risk to develop type 2 diabetes.


Subject(s)
Blood Glucose/metabolism , Diet, Protein-Restricted , Gene Expression Regulation/physiology , Homeodomain Proteins/metabolism , Maternal Nutritional Physiological Phenomena/physiology , Pancreas/metabolism , Prenatal Exposure Delayed Effects/metabolism , Trans-Activators/metabolism , Animals , Female , Glucokinase/genetics , Glucokinase/metabolism , Glucose Tolerance Test , Glucose Transporter Type 2/genetics , Glucose Transporter Type 2/metabolism , Homeodomain Proteins/genetics , Homeostasis/physiology , Insulin/metabolism , Male , Pregnancy , Prenatal Exposure Delayed Effects/genetics , Rats , Rats, Wistar , Trans-Activators/genetics
19.
J Mol Endocrinol ; 53(2): 237-46, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25122159

ABSTRACT

In maternal diabetes, the fetal heart can be structurally and functionally affected. Maternal diets enriched in certain unsaturated fatty acids can activate the nuclear receptors peroxisome proliferator-activated receptors (PPARs) and regulate metabolic and anti-inflammatory pathways during development. Our aim was to investigate whether PPARα expression, lipid metabolism, lipoperoxidation, and nitric oxide (NO) production are altered in the fetal hearts of diabetic rats, and to analyze the putative effects of in vivo PPAR activation on these parameters. We found decreased PPARα expression in the hearts of male but not female fetuses of diabetic rats when compared with controls. Fetal treatments with the PPARα ligand leukotriene B4 upregulated the expression of PPARα and target genes involved in fatty acid oxidation in the fetal hearts. Increased concentrations of triglycerides, cholesterol, and phospholipids were found in the hearts of fetuses of diabetic rats. Maternal treatments with diets supplemented with 6% olive oil or 6% safflower oil, enriched in unsaturated fatty acids that can activate PPARs, led to few changes in lipid concentrations, but up-regulated PPARα expression in fetal hearts. NO production, which was increased in the hearts of male and female fetuses in the diabetic group, and lipoperoxidation, which was increased in the hearts of male fetuses in the diabetic group, was reduced by the maternal treatments supplemented with safflower oil. In conclusion, impaired PPARα expression, altered lipid metabolism, and increased oxidative and nitridergic pathways were evidenced in hearts of fetuses of diabetic rats and were regulated in a gender-dependent manner by treatments enriched with PPAR ligands.


Subject(s)
Diabetes Mellitus, Experimental/metabolism , Fetal Heart/metabolism , Metabolic Networks and Pathways , Myocardium/metabolism , Peroxisome Proliferator-Activated Receptors/metabolism , Animals , Diet , Female , Ligands , Lipid Metabolism , Lipid Peroxidation , Male , Nitric Oxide/metabolism , Olive Oil , Oxidation-Reduction , PPAR alpha/agonists , PPAR alpha/genetics , PPAR alpha/metabolism , Peroxisome Proliferator-Activated Receptors/agonists , Peroxisome Proliferator-Activated Receptors/genetics , Plant Oils/administration & dosage , Pregnancy , Rats , Safflower Oil/administration & dosage
20.
Exp Biol Med (Maywood) ; 238(10): 1147-59, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23986224

ABSTRACT

Low birth weight and poor foetal growth following low protein (LP) exposure are associated with altered islet development and glucose intolerance in adulthood. Additionally, LP-fed offspring fail to regenerate their ß-cells following depletion with streptozotocin (STZ) in contrast to control-fed offspring that restore ß-cell mass. Our objective was to identify signalling pathways and cellular functions that may be critically altered in LP offspring rendering them susceptible to developing long-term glucose intolerance and decreased ß-cell plasticity. Pregnant Balb/c mice were fed a control (C; 20% protein) or an isocaloric LP (8% protein) diet throughout gestation and C diet thereafter. Female offspring were injected intraperitoneally with 35 mg/kg STZ or vehicle on days 1 to 5 for each dietary treatment. At 30 days of age, total RNA was extracted from pancreatic tissue for microarray analysis using the Affymetrix GeneChip Mouse Genome 430 2.0. Gene and protein expression were quantified from isolated islets. Finally, ß-cell proliferation was determined in vitro following REG1α treatment. The microarray data and GO enrichment analysis indicated that foetal protein restriction alters the early expression of genes necessary for many cell functions, such as oxidative phosphorylation and free radical scavenging. Expression of Reg1 was upregulated following STZ, whereas protein content was decreased in LP + STZ islets. Furthermore, REG1α failed to stimulate ß-cell proliferation in vitro in LP + STZ islets. Therefore, early nutritional insults may programme the Reg1 pathway resulting in a limited ability to increase ß-cell mass during metabolic stress. In conclusion, this study implicates the Reg1 pathway in ß-cell regeneration and describes altered programming of gene expression in LP offspring, which underlies later development of cell dysfunction and glucose intolerance in adulthood.


Subject(s)
Cell Proliferation , Glucose Intolerance/physiopathology , Insulin-Secreting Cells/physiology , Lithostathine/metabolism , Protein Deficiency/complications , Regeneration , Siblings , Animals , Female , Gene Expression Profiling , Lithostathine/administration & dosage , Mice , Mice, Inbred BALB C , Pregnancy , Proteins , Proteome/analysis , Streptozocin/administration & dosage , Streptozocin/toxicity
SELECTION OF CITATIONS
SEARCH DETAIL
...