Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
Mais filtros

Bases de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Int J Mol Sci ; 22(23)2021 Nov 29.
Artigo em Inglês | MEDLINE | ID: mdl-34884717

RESUMO

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.


Assuntos
Acetona/farmacologia , Antimaláricos/uso terapêutico , Artesunato/uso terapêutico , Diabetes Gestacional/tratamento farmacológico , Pâncreas/efeitos dos fármacos , Animais , Apelina/metabolismo , Modelos Animais de Doenças , Avaliação Pré-Clínica de Medicamentos , Jejum , Feminino , Intestinos/efeitos dos fármacos , Placenta/efeitos dos fármacos , Placenta/metabolismo , Gravidez , Resultado da Gravidez
2.
Cell Mol Neurobiol ; 40(3): 369-380, 2020 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-31522299

RESUMO

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.


Assuntos
Doença de Alzheimer/complicações , Doença de Alzheimer/patologia , Isquemia Encefálica/complicações , Isquemia Encefálica/patologia , Circulação Cerebrovascular/fisiologia , Neuroglia/fisiologia , Doença de Alzheimer/induzido quimicamente , Doença de Alzheimer/fisiopatologia , Peptídeos beta-Amiloides , Animais , Astrócitos/patologia , Astrócitos/fisiologia , Encéfalo/irrigação sanguínea , Encéfalo/patologia , Encéfalo/fisiopatologia , Isquemia Encefálica/fisiopatologia , Modelos Animais de Doenças , Masculino , Neurônios/patologia , Neurônios/fisiologia , Ratos , Ratos Wistar , Traumatismo por Reperfusão/complicações , Traumatismo por Reperfusão/patologia , Traumatismo por Reperfusão/fisiopatologia
3.
Cell Mol Neurobiol ; 40(3): 381-382, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-31599387

RESUMO

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.

4.
J Biol Chem ; 293(44): 17218-17228, 2018 11 02.
Artigo em Inglês | MEDLINE | ID: mdl-30217817

RESUMO

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.


Assuntos
Citocinas/imunologia , Diabetes Mellitus/genética , Células Secretoras de Insulina/imunologia , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos/genética , Receptor Tipo 5 de Fator de Crescimento de Fibroblastos/genética , Animais , Diabetes Mellitus/imunologia , Dimerização , Feminino , Fator 2 de Crescimento de Fibroblastos/imunologia , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos NOD , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos/química , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos/imunologia , Receptor Tipo 5 de Fator de Crescimento de Fibroblastos/química , Receptor Tipo 5 de Fator de Crescimento de Fibroblastos/imunologia , Regulação para Cima
5.
Reproduction ; 149(3): 293-303, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25667428

RESUMO

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.


Assuntos
Glicemia/metabolismo , Dieta com Restrição de Proteínas , Regulação da Expressão Gênica/fisiologia , Proteínas de Homeodomínio/metabolismo , Fenômenos Fisiológicos da Nutrição Materna/fisiologia , Pâncreas/metabolismo , Efeitos Tardios da Exposição Pré-Natal/metabolismo , Transativadores/metabolismo , Animais , Feminino , Glucoquinase/genética , Glucoquinase/metabolismo , Teste de Tolerância a Glucose , Transportador de Glucose Tipo 2/genética , Transportador de Glucose Tipo 2/metabolismo , Proteínas de Homeodomínio/genética , Homeostase/fisiologia , Insulina/metabolismo , Masculino , Gravidez , Efeitos Tardios da Exposição Pré-Natal/genética , Ratos , Ratos Wistar , Transativadores/genética
6.
J Endocrinol ; 260(1)2024 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-37855335

RESUMO

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.


Assuntos
Canabidiol , Intolerância à Glucose , Resistência à Insulina , Células Secretoras de Insulina , Gravidez , Ratos , Feminino , Masculino , Humanos , Animais , Lactente , Canabidiol/toxicidade , Intolerância à Glucose/induzido quimicamente , Ratos Wistar
7.
Am J Physiol Endocrinol Metab ; 304(10): E1064-76, 2013 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-23531612

RESUMO

Adult mice lacking functional GABAB receptors (GABAB1KO) have glucose metabolism alterations. Since GABAB receptors (GABABRs) are expressed in progenitor cells, we evaluated islet development in GABAB1KO mice. Postnatal day 4 (PND4) and adult, male and female, GABAB1KO, and wild-type littermates (WT) were weighed and euthanized, and serum insulin and glucagon was measured. Pancreatic glucagon and insulin content were assessed, and pancreas insulin, glucagon, PCNA, and GAD65/67 were determined by immunohistochemistry. RNA from PND4 pancreata and adult isolated islets was obtained, and Ins1, Ins2, Gcg, Sst, Ppy, Nes, Pdx1, and Gad1 transcription levels were determined by quantitative PCR. The main results were as follows: 1) insulin content was increased in PND4 GABAB1KO females and in both sexes in adult GABAB1KOs; 2) GABAB1KO females had more clusters (<500 µm(2)) and less islets than WT females; 3) cluster proliferation was decreased at PND4 and increased in adult GABAB1KO mice; 4) increased ß-area at the expense of the α-cell area was present in GABAB1KO islets; 5) Ins2, Sst, and Ppy transcription were decreased in PND4 GABAB1KO pancreata, adult GABAB1KO female islets showed increased Ins1, Ins2, and Sst expression, Pdx1 was increased in male and female GABAB1KO islets; and 6) GAD65/67 was increased in adult GABAB1KO pancreata. We demonstrate that several islet parameters are altered in GABAB1KO mice, further pinpointing the importance of GABABRs in islet physiology. Some changes persist from neonatal ages to adulthood (e.g., insulin content in GABAB1KO females), whereas other features are differentially regulated according to age (e.g., Ins2 was reduced in PND4, whereas it was upregulated in adult GABAB1KO females).


Assuntos
Resistência à Insulina/fisiologia , Ilhotas Pancreáticas/fisiologia , Receptores de GABA-B/deficiência , Animais , Animais Recém-Nascidos , Peso Corporal/fisiologia , Feminino , Regulação da Expressão Gênica , Glucagon/sangue , Glucagon/genética , Glucagon/fisiologia , Glutamato Descarboxilase/fisiologia , Insulina/sangue , Insulina/genética , Insulina/fisiologia , Ilhotas Pancreáticas/crescimento & desenvolvimento , Ilhotas Pancreáticas/metabolismo , Ilhotas Pancreáticas/ultraestrutura , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Tamanho do Órgão/fisiologia , Antígeno Nuclear de Célula em Proliferação/análise , Antígeno Nuclear de Célula em Proliferação/fisiologia , RNA/química , RNA/genética , Reação em Cadeia da Polimerase em Tempo Real
8.
Reproduction ; 145(6): 609-20, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23613616

RESUMO

Dietary protein restriction during pregnancy and lactation in rats impairs ß-cell function and mass in neonates and leads to glucose intolerance in adult offspring. Maternal taurine (Tau) supplementation during pregnancy in rats restores ß-cell function and mass in neonates, but its long-term effects are unclear. The prevention of postnatal catch-up growth has been suggested to improve glucose tolerance in adult offspring of low-protein (LP)-fed mothers. The objective of this study was to examine the relative contribution of ß-cell dysfunction and insulin resistance to impaired glucose tolerance in 130-day-old rat offspring of LP-fed mothers and the effects of maternal Tau supplementation on ß-cell function and insulin resistance in these offspring. Pregnant rats were fed i) control, ii) LP, and iii) LP+Tau diets during gestation and lactation. Offspring were given a control diet following weaning. A fourth group consisting of offspring of LP-fed mothers, maintained on a LP diet following weaning, was also studied (LP-all life). Insulin sensitivity in the offspring of LP-fed mothers was reduced in females but not in males. In both genders, LP exposure decreased ß-cell function. Tau supplementation improved insulin sensitivity in females and ß-cell function in males. The LP-all life diet improved ß-cell function in males. We conclude that i) maternal Tau supplementation has persistent effects on improving glucose metabolism (ß-cell function and insulin sensitivity) in adult rat offspring of LP-fed mothers and ii) increasing the amount of protein in the diet of offspring adapted to a LP diet after weaning may impair glucose metabolism (ß-cell function) in a gender-specific manner.


Assuntos
Diabetes Mellitus Tipo 2/prevenção & controle , Dieta com Restrição de Proteínas/efeitos adversos , Suplementos Nutricionais , Resistência à Insulina , Células Secretoras de Insulina/metabolismo , Fenômenos Fisiológicos da Nutrição Materna , Taurina/uso terapêutico , Animais , Peptídeo C/sangue , Diabetes Mellitus Tipo 2/etiologia , Diabetes Mellitus Tipo 2/imunologia , Diabetes Mellitus Tipo 2/patologia , Proteínas Alimentares/efeitos adversos , Feminino , Células Secretoras de Insulina/imunologia , Células Secretoras de Insulina/patologia , Lactação , Masculino , Gravidez , Complicações na Gravidez/etiologia , Complicações na Gravidez/fisiopatologia , Deficiência de Proteína/etiologia , Deficiência de Proteína/fisiopatologia , Ratos , Ratos Wistar , Caracteres Sexuais , Desmame
9.
Can J Diabetes ; 46(8): 851-862, 2022 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-35985923

RESUMO

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.


Assuntos
Intolerância à Glucose , Resistência à Insulina , Efeitos Tardios da Exposição Pré-Natal , Animais , Feminino , Masculino , Gravidez , Ratos , Dronabinol/efeitos adversos , Glucagon , Insulina , Efeitos Tardios da Exposição Pré-Natal/induzido quimicamente , Ratos Wistar , Estatmina
10.
J Comp Neurol ; 529(2): 421-433, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-32447764

RESUMO

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.


Assuntos
Lesões Encefálicas/patologia , Sobrevivência Celular/fisiologia , Córtex Cerebral/patologia , Córtex Cerebral/fisiologia , Degeneração Neural/patologia , Animais , Córtex Cerebral/química , Corpo Estriado/química , Corpo Estriado/patologia , Corpo Estriado/fisiologia , Masculino , Ratos , Ratos Wistar
11.
J Dev Orig Health Dis ; 12(6): 915-922, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-33353580

RESUMO

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.


Assuntos
Dieta com Restrição de Proteínas/efeitos adversos , Sepse/fisiopatologia , Fenômenos Fisiológicos da Nutrição Animal , Animais , Dieta com Restrição de Proteínas/métodos , Modelos Animais de Doenças , Quebeque , Ratos , Ratos Wistar , Sepse/dietoterapia
12.
Pediatr Res ; 68(1): 16-22, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20386490

RESUMO

Beta cells are partially replaced in neonatal rodents after deletion with streptozotocin (STZ). Exposure of pregnant rats to a low protein (LP) diet impairs endocrine pancreas development in the offspring, leading to glucose intolerance in adulthood. Our objective was to determine whether protein restriction has a similar effect on the offspring in mice, and if this alters the capacity for beta cell regeneration after STZ. Pregnant Balb/c mice were fed a control (C) (20% protein) or an isocaloric LP (8% protein) diet during gestation. Pups were given 35 mg/kg STZ (or vehicle) from d 1 to 5 for each dietary treatment. Histologic analysis showed that C-fed offspring had largely replaced beta cell mass (BCM) after STZ by d 30, but this was not sustained over time. Female LP-fed offspring showed an initial increase in BCM by d 14 but developed glucose intolerance by d 130. In contrast, male LP offspring showed no changes in BCM or glucose tolerance. However, LP exposure limited the capacity for recovery of BCM in both genders after STZ treatment.


Assuntos
Proteínas Alimentares/metabolismo , Células Secretoras de Insulina/fisiologia , Pâncreas/citologia , Pâncreas/embriologia , Pâncreas/crescimento & desenvolvimento , Regeneração/fisiologia , Animais , Glicemia/metabolismo , Peso Corporal , Diabetes Mellitus Experimental/metabolismo , Dieta , Feminino , Humanos , Insulina/metabolismo , Células Secretoras de Insulina/citologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Tamanho do Órgão , Pâncreas/metabolismo , Gravidez , Distribuição Aleatória , Ratos
13.
J Endocrinol ; 246(2): 175-187, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32485676

RESUMO

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.


Assuntos
Diabetes Mellitus Experimental/dietoterapia , Diabetes Gestacional/dietoterapia , Azeite de Oliva/uso terapêutico , Animais , Suplementos Nutricionais , Feminino , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Leucotrieno B4/farmacologia , Metabolismo dos Lipídeos/efeitos dos fármacos , Masculino , Ácido Oleico/uso terapêutico , Receptores Ativados por Proliferador de Peroxissomo/agonistas , Gravidez , Ratos , Estreptozocina/toxicidade , Transativadores/genética , Transativadores/metabolismo
14.
J Comp Neurol ; 528(1): 48-60, 2020 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-31265125

RESUMO

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.


Assuntos
Peptídeos beta-Amiloides/toxicidade , Encéfalo/patologia , Infarto Cerebral/patologia , Endotelina-1/toxicidade , Rede Nervosa/patologia , Peptídeos beta-Amiloides/administração & dosagem , Animais , Encéfalo/anatomia & histologia , Encéfalo/efeitos dos fármacos , Infarto Cerebral/induzido quimicamente , Endotelina-1/administração & dosagem , Injeções Intraventriculares , Masculino , Rede Nervosa/anatomia & histologia , Rede Nervosa/efeitos dos fármacos , Ratos , Ratos Wistar
15.
Reprod Toxicol ; 94: 84-91, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32325173

RESUMO

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.


Assuntos
Dronabinol/toxicidade , Glucose/metabolismo , Ilhotas Pancreáticas/efeitos dos fármacos , Efeitos Tardios da Exposição Pré-Natal , Animais , Feminino , Homeostase/efeitos dos fármacos , Insulina/metabolismo , Ilhotas Pancreáticas/crescimento & desenvolvimento , Masculino , Troca Materno-Fetal , Gravidez , Ratos Wistar , Caracteres Sexuais
16.
FASEB J ; 22(7): 2452-64, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-18323405

RESUMO

Neuropeptide Y (NPY) is synthesized in neural tissue of the central and peripheral nervous systems and has a number of important functions besides regulating appetite and energy homeostasis. Here we identify a novel site of NPY biosynthesis and a role for NPY in promoting proliferation of adipocyte precursor cells. We show that NPY mRNA is not only expressed in visceral adipose tissue (VAT) but that its levels are up-regulated 6-fold in our early-life programmed rat model of increased visceral adiposity. This is accompanied by a parallel rise in NPY protein, demonstrating that VAT is a novel peripheral site of NPY biosynthesis. Furthermore, NPY mRNA expression is also elevated >2-fold in VAT of obese Zucker rats. Importantly, NPY stimulates proliferation of primary rat preadipocytes as well as 3T3-L1 preadipocytes in vitro. This mitogenic effect appears to be mediated by the Y1 receptor and involves the activation of extracellular related kinase 1/2. In addition, insulin and glucocorticoid up-regulate VAT NPY expression in lean but not obese Zucker rats. Taken together, these results suggest that an enhanced local expression of NPY within VAT may be a common feature of and contribute to the molecular mechanisms underlying increased visceral adiposity.


Assuntos
Adipócitos/química , Tecido Adiposo/fisiologia , Neuropeptídeo Y/fisiologia , Receptores de Neuropeptídeo Y/fisiologia , Animais , Divisão Celular , Feminino , Tamanho da Ninhada de Vivíparos , Masculino , Modelos Animais , Neuropeptídeo Y/biossíntese , Neuropeptídeo Y/genética , Gravidez , Ratos , Ratos Wistar , Ratos Zucker , Vísceras
17.
Mol Neurobiol ; 56(4): 2714-2727, 2019 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-30054859

RESUMO

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.


Assuntos
Peptídeos beta-Amiloides/toxicidade , Corpo Estriado/patologia , Hipocampo/irrigação sanguínea , Hipocampo/lesões , Hipocampo/patologia , Animais , Corpo Estriado/efeitos dos fármacos , Hipocampo/metabolismo , Masculino , Ratos Wistar
18.
Sci Rep ; 8(1): 5136, 2018 03 23.
Artigo em Inglês | MEDLINE | ID: mdl-29572520

RESUMO

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.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Isquemia Encefálica/metabolismo , Transtornos Cognitivos/metabolismo , Corpo Estriado/metabolismo , Fator de Crescimento Insulin-Like I/metabolismo , Insulina/metabolismo , Transtornos da Memória/metabolismo , Transdução de Sinais , Animais , Isquemia Encefálica/patologia , Transtornos Cognitivos/patologia , Comorbidade , Corpo Estriado/patologia , Modelos Animais de Doenças , Proteínas Substratos do Receptor de Insulina/metabolismo , Masculino , Transtornos da Memória/patologia , Ratos , Ratos Wistar
19.
Islets ; 10(4): 137-150, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30110202

RESUMO

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.


Assuntos
Células-Tronco Hematopoéticas/fisiologia , Hiperglicemia/terapia , Insulina/sangue , Células-Tronco Mesenquimais/fisiologia , Transplante de Células-Tronco/métodos , Animais , Glicemia/metabolismo , Medula Óssea , Modelos Animais de Doenças , Teste de Tolerância a Glucose/métodos , Células Secretoras de Insulina/patologia , Células Secretoras de Insulina/fisiologia , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Fatores de Tempo , Resultado do Tratamento
20.
J Endocr Soc ; 2(2): 178-189, 2018 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-29450407

RESUMO

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.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA